1
|
Fang W, Ma X, Liu B. Global research progress in antibody-drug conjugates for solid tumors: Bibliometrics and visualized analysis. Hum Vaccin Immunother 2025; 21:2472493. [PMID: 40013384 PMCID: PMC11869778 DOI: 10.1080/21645515.2025.2472493] [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: 09/22/2024] [Revised: 02/14/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025] Open
Abstract
Recently, the use of antibody-drug conjugates (ADCs) in the research and management of solid tumors has increased, making them a key focus in the field of oncology. In this study, we performed a comprehensive literature review of ADCs use in solid tumor treatment. We retrieved data from the Web of Science Core Collection (WoSCC). Following literature retrieval, we conducted a thorough bibliometric and knowledge-mapping analysis of the collected articles. There was a rapid growth in the number of annual publications in this field. The United States had the highest publication volumes and led ADC research for solid tumors. Additionally, The Dana-Farber Cancer Institute had the highest output, and G. Curigliano was identified as the most productive author. The journal "Cancers" led in the publishing of ADC research on solid tumors. Furthermore, key clustering terms such as "breast cancer," "targeted therapy," "bladder cancer," "ovarian cancer," "expression," and "drug delivery" emerged in this field as the research progressed. We identified six key themes by literature co-citation analysis, involving the research on the application of four ADCs in breast cancer, as well as the analysis of ADCs design, mechanisms, and strategies for reducing cytotoxicity. At the same time, based on the analysis of papers that have experienced a citation burst recently, we explored the future development trends of this field. Overall, our inaugural bibliometric analysis of ADCs for solid tumor research provides a systematic framework to guide future studies in this field. Therefore, facilitating and promoting further development in this area.
Collapse
Affiliation(s)
- Wenjun Fang
- Department of Pharmacy, The Affiliated Yancheng Maternity & Child Health Hospital of Yangzhou University, Yancheng, China
| | - Xueqing Ma
- Department of Dermatology, The Affiliated Yancheng Maternity & Child Health Hospital of Yangzhou University, Yancheng, China
| | - Ben Liu
- Yancheng No.1 People’s Hospital, Affiliated Hospital of Medical School, Nanjing University, Yancheng, China
- Pediatric Intensive Care Unit, The First People’s Hospital of Yancheng, Yancheng, China
| |
Collapse
|
2
|
Wang Y, Tang C, Wang K, Zhang X, Zhang L, Xiao X, Lin H, Xiong L. The role of ferroptosis in breast cancer: Tumor progression, immune microenvironment interactions and therapeutic interventions. Eur J Pharmacol 2025; 996:177561. [PMID: 40154567 DOI: 10.1016/j.ejphar.2025.177561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Ferroptosis represents a distinctive and distinct form of regulated cellular death, which is driven by the accumulation of lipid peroxidation. It is distinguished by altered redox lipid metabolism and is linked to a spectrum of cellular activities, including cancer. In breast cancer (BC), with triple negative breast cancer (TNBC) being an iron-and lipid-rich tumor, inducing ferroptosis was thought to be a novel approach to killing breast tumor cells. However, in the recent past, a novel conceptual framework has emerged which posits that in addition to the promotion of tumor cell death, ferritin deposition has a potent immunosuppressive effect on the tumor immune microenvironment (TIME) via the influence on both innate and adaptive immune responses. TIME of BC includes various cell populations from both the innate and adaptive immune systems. In this review, the internal association between iron homeostasis and the progression of ferroptosis, along with the common inducers and protectors of ferroptosis in BC, are discussed in detail. Furthermore, a comprehensive analysis is conducted on the dual role of ferroptosis in immune cells and proto-oncogenic functions, along with an evaluation of the potential applications of immunogenic cell death-targeted immunotherapy in TIME of BC. It is anticipated that our review will inform future research endeavors that seek to integrate ferroptosis and immunotherapy in the management of BC.
Collapse
Affiliation(s)
- Yi Wang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Chuanyun Tang
- First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Keqin Wang
- First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiaoan Zhang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lifang Zhang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xinghua Xiao
- Department of Pathology, The First Affiliated Hospital, Nanchang University, 17 Yongwaizheng Road, Nanschang, 330066, China
| | - Hui Lin
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lixia Xiong
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
3
|
Kydd AR, Sarwar MS, Atiq S, Chelluri R, Gurram S, Chandran E, Simon N, Stukes I, Weng S, Yousefi-Rad A, Banday AR, Boudjadi S, Apolo AB. Antibody-drug conjugates in rare genitourinary tumors: review and perspectives. Curr Opin Oncol 2025; 37:250-258. [PMID: 40110990 PMCID: PMC11970986 DOI: 10.1097/cco.0000000000001141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
PURPOSE OF REVIEW Rare cancers of the genitourinary (GU) tract are often clinically aggressive yet have few or no standard-of-care treatments. Multiple antibody-drug conjugates (ADCs) have been approved in solid malignancies. This review explores the use of ADCs in rare GU tumors in the context of biological pathways and ongoing research in solid tumors. RECENT FINDINGS Few clinical trials of ADCs focus on recruiting participants with rare tumors of the GU tract, including trials testing enfortumab vedotin as monotherapy or combined with pembrolizumab, and sacituzumab govitecan as monotherapy or combined with atezolizumab. We highlight many ongoing trials of novel ADCs for advanced/metastatic solid tumors and emphasize the potential eligibility of patients with rare GU tumors for tumor-agnostic trials. SUMMARY ADCs are being tested in multiple solid tumors, including rare GU tumors. Ongoing preclinical research supports the use of some ADCs in several rare GU tumors and improves our understanding of their pathophysiology.
Collapse
Affiliation(s)
- Andre R. Kydd
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Md. Shahid Sarwar
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali-3814, Bangladesh
| | - Saad Atiq
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Raju Chelluri
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sandeep Gurram
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elias Chandran
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nicholas Simon
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ian Stukes
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sally Weng
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Abbas Yousefi-Rad
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - A. Rouf Banday
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Salah Boudjadi
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrea B. Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Luz P, Ramos S, Oliveira MJ, Costa JG, Saraiva N, Fernandes AS. Interaction between redox regulation, immune activation, and response to treatment in HER2+ breast cancer. Redox Biol 2025; 82:103609. [PMID: 40174475 PMCID: PMC11999322 DOI: 10.1016/j.redox.2025.103609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/04/2025] Open
Abstract
In HER2+ breast cancer (BC), neoadjuvant therapy represents an ideal scenario for translational research, considering pathological complete response (pCR) as an endpoint. In these patients, achieving pCR after neoadjuvant therapy is associated with a better prognosis. However, biomarkers are needed to tailor optimal treatment for each patient. Evaluating tumour-infiltrating lymphocytes (TILs) has gained attention in predicting pCR. In the context of metastatic disease, TILs also appear to play a role in predicting outcomes. The interaction between the presence of TILs and reactive oxygen species (ROS) remains an area to be explored. ROS are critical for tumour cell homeostasis, and different levels can trigger differential biological responses in cancer cells and their microenvironment. Nevertheless, the influence of ROS on treatment efficacy and prognosis in patients with HER2+ BC remains to be elucidated. In this article, we reviewed the interplay between treatment response, immune system activation, and ROS production in HER2+ BC and suggested novel areas of intervention and research. We also present a bioinformatic analysis demonstrating that the altered expression of several redox-related genes could be associated with the prevalence of immune cell populations in the tumour microenvironment and with patient survival. New biomarkers are thus suggested and should be further explored to tailor the best treatment to each patient.
Collapse
Affiliation(s)
- Paulo Luz
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Lisbon, Portugal; Universidad de Alcalá de Henares. Departamento de Ciencias Biomédicas, Alcalá de Henares, Madrid, Spain; Medical Oncology Department, Unidade Local de Saúde do Baixo Alentejo - Hospital José Joaquim Fernandes, Beja, Portugal
| | - Sofia Ramos
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Lisbon, Portugal; Universidad de Alcalá de Henares. Departamento de Ciencias Biomédicas, Alcalá de Henares, Madrid, Spain
| | - Maria José Oliveira
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - João G Costa
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Lisbon, Portugal
| | - Nuno Saraiva
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Lisbon, Portugal
| | - Ana S Fernandes
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Lisbon, Portugal.
| |
Collapse
|
5
|
Xiong S, Huang Z, Mukwaya V, Zhao W, Wang L, Dou H. Cell-Targeting Bio-Catalytic Killer Protocell for High-Order Assembly Guided Cancer Cell Inhibition. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2500047. [PMID: 40270292 DOI: 10.1002/smll.202500047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/03/2025] [Indexed: 04/25/2025]
Abstract
The design and construction of synthetic therapeutic protocells capable of engaging in high-order assembly with living cells represent a significant challenge in synthetic biology and bioengineering. Inspired by cell membrane receptor-ligand systems, a protocell bioreactor is developed for targeted cancer cell elimination. This is achieved by constructing orthogonal, polysaccharide-based protocells (polysaccharidosomes, P-somes) through a bottom-up approach that leverages host-guest chemistry. The protocells are assembled via electrostatically-driven self-assembly of β-cyclodextrin (β-CD)-modified amino-dextran on a sacrificial template encapsulating glucose oxidase (GOx). To enable specific cancer cell targeting and catalytic activity, cell-targeting ligands (arginylglycylaspartic acid, cRGD) and catalase-like platinum-gold nanoparticles (Pt-AuNPs) are introduced through host-guest interactions, forming a fully functional, cell-targeting, bio-catalytic killer protocell. These protocells are programmed to spatially couple the GOx/Pt-AuNP catalytic reaction cascade. In the presence of glucose and hydroxyurea, this cascade generates a localized flux of nitric oxide (NO), which is exploited for in vitro cancer cell inhibition. Overall, the results highlight the potential of integrating orthogonal and synergistic tumor inhibition mechanisms within synthetic microcompartments. This platform demonstrates promise for future therapeutic applications, especially in cancer treatment, and represents a step forward in the development of programmable protocell-based therapeutic systems.
Collapse
Affiliation(s)
- Shuhan Xiong
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zeqi Huang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Vincent Mukwaya
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Weili Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Li Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongjing Dou
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
6
|
Singh R, Patel K, Xu H, Adeniyi A, Upshaw JN, Van Buren P, Kaufman PA, Dittus K, Landry KK. Cardio-Oncology and Breast Cancer Therapies. Curr Treat Options Oncol 2025:10.1007/s11864-025-01311-x. [PMID: 40257669 DOI: 10.1007/s11864-025-01311-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/22/2025]
Abstract
OPINION STATEMENT Assessing cardiac risk prior to initiating breast cancer treatment, monitoring cardiac function during treatment, and implementing appropriate follow-up strategies are essential components of managing cardiotoxicity in breast cancer patients. A comprehensive cardiovascular evaluation should be conducted before treatment, including a detailed medical history, physical examination, and baseline cardiac imaging. Risk stratification tools can aid in determining the individual patient's risk profile. Close monitoring of cardiac function, including regular assessment of left ventricular ejection fraction (LVEF) and monitoring for signs and symptoms of cardiac dysfunction, is crucial during treatment. Prompt action should be taken if an adverse cardiovascular event is detected, including considering discontinuing or modifying the treatment regimen. Appropriate follow-up care is essential to monitor for long-term cardiac effects and optimize cardiovascular health in breast cancer survivors. Regular cardiovascular assessments, lifestyle modifications, and collaboration between healthcare professionals are important in managing cardiotoxicity effectively.
Collapse
Affiliation(s)
- Rohit Singh
- Division of Hematology/Oncology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- University of Vermont Cancer Center, Burlington, VT, USA
| | - Krina Patel
- Division of Cardiology, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Haze Xu
- Division of Hematology/Oncology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- University of Vermont Cancer Center, Burlington, VT, USA
| | - Aderonke Adeniyi
- Division of Cardiology, Department of Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Jenica N Upshaw
- Division of Cardiology, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Peter Van Buren
- Division of Cardiology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Peter A Kaufman
- Division of Hematology/Oncology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- University of Vermont Cancer Center, Burlington, VT, USA
| | - Kim Dittus
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Kara K Landry
- Division of Hematology/Oncology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
- University of Vermont Cancer Center, Burlington, VT, USA.
| |
Collapse
|
7
|
Li C, Dai H, Guo X, Zhou L, Jiang M. Comprehensive review of non-invasive-treatment-related cardiovascular toxicity in breast cancer. iScience 2025; 28:111759. [PMID: 40207253 PMCID: PMC11980005 DOI: 10.1016/j.isci.2025.111759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025] Open
Abstract
Cardiovascular toxicity is a significant side effect of breast cancer treatment and has emerged as a leading cause of non-tumor-related deaths among breast cancer survivors, emphasizing the critical need for effective monitoring and management of these complications. As breast cancer remains the most prevalent cancer among women, advancements in survival rates have been achieved through treatments such as chemotherapy, targeted therapy, endocrine therapy, immunotherapy, and radiotherapy. This review provides a comprehensive understanding of the cardiovascular toxicity mechanisms associated with both established and emerging breast cancer therapies, identifies potential therapeutic targets and monitoring strategies, and highlights key deficiencies and challenges in the field. By offering insights into the early detection, prevention, and management of cardiovascular complications, this review aims to guide future research directions and clinical practices, ultimately improving outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Cenyu Li
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Huijuan Dai
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xinning Guo
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Liheng Zhou
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Meng Jiang
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| |
Collapse
|
8
|
Lee HK, Kim B, Ko YG, Chung SW, Shim WS, Choi SY, Lee SR, Kim SY, Byun Y. Enhancing the bystander effect of antibody-drug conjugate by using a novel caspase-3 cleavable peptide linker to overcome tumor heterogeneity. J Control Release 2025; 382:113738. [PMID: 40246243 DOI: 10.1016/j.jconrel.2025.113738] [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: 11/18/2024] [Revised: 03/26/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025]
Abstract
Tumor heterogeneity is a major obstacle to effective targeted therapies, including those utilizing antibody-drug conjugates (ADCs). Although some ADCs employ the bystander effect to eliminate neighboring antigen-negative cells, their efficacy often diminishes as antigen-positive cell populations decrease within heterogeneous tumors. To address this limitation in ADC therapies, we developed a novel ADC using a caspase substrate, Asp-Glu-Val-Asp (DEVD), as a linker to generate a more potent and sustained bystander effect. The DEVD ADC effectively targeted antigen-positive cells and released its payload via cathepsin B cleavage. Notably, it exhibited a significant bystander effect mediated by the caspase-3-triggered extracellular cleavage of the linker, enhancing payload release into the tumor microenvironment. In breast cancer xenograft models, the DEVD ADC maintained its efficacy and continued to exert a bystander effect even after the depletion of antigen-positive cells, thereby overcoming challenges posed by tumor heterogeneity. These findings emphasize the potential of DEVD linkers in enhancing ADC efficacy against heterogeneous solid tumors, offering a promising strategy to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Ha Kyeong Lee
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergent Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Byoungmo Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Yoon Gun Ko
- Pharosgen Co. Ltd, 2-404 Jangji-dong 892, Seoul 05852, Republic of Korea
| | - Seung Woo Chung
- Center for Nanomedicine, Wilmer Eye Institute and Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Wan Seob Shim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - So-Young Choi
- New Drug Development Center Osong Medical Innovation Foundation 123 Osongsaengmyeong-ro, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Se-Ra Lee
- New Drug Development Center Osong Medical Innovation Foundation 123 Osongsaengmyeong-ro, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Sang Yoon Kim
- Pharosgen Co. Ltd, 2-404 Jangji-dong 892, Seoul 05852, Republic of Korea.
| | - Youngro Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
9
|
Hong Y, He J, Deng D, Liu Q, Zu X, Shen Y. Targeting kinases that regulate programmed cell death: a new therapeutic strategy for breast cancer. J Transl Med 2025; 23:439. [PMID: 40229646 PMCID: PMC11995514 DOI: 10.1186/s12967-025-06367-9] [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: 12/09/2024] [Accepted: 03/08/2025] [Indexed: 04/16/2025] Open
Abstract
Breast cancer is one of the most prevalent malignant tumors among women and ranks as the second leading cause of cancer-related deaths in females, primarily due to delays in diagnosis and shortcomings in treatment strategies. Consequently, there is a pressing need to identify reliable therapeutic targets and strategies. In recent years, the identification of effective biomarkers-particularly novel molecular therapeutic targets-has become a focal point in breast cancer research, aimed at predicting disease aggressiveness and monitoring treatment responses. Simultaneously, advancements in understanding the molecular mechanisms underlying cellular programmed death have opened new avenues for targeting kinase-regulated programmed cell death as a viable therapeutic strategy. This review summarizes the latest research progress regarding kinase-regulated programmed death (including apoptosis, pyroptosis, autophagy, necroptosis, and ferroptosis) in breast cancer treatment. It covers the key kinases involved in this mechanism, their roles in the onset and progression of breast cancer, and strategies for modulating these kinases through pharmacological interventions.
Collapse
Affiliation(s)
- Yun Hong
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hunan Province Clinical Research Center for Accurate Diagnosis and Treatment of High-Incidence Sexually Transmitted Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jun He
- Department of Spine Surgery, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, China
| | - Dan Deng
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hunan Province Clinical Research Center for Accurate Diagnosis and Treatment of High-Incidence Sexually Transmitted Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qinyue Liu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hunan Province Clinical Research Center for Accurate Diagnosis and Treatment of High-Incidence Sexually Transmitted Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
- Hunan Provincial Clinical Medical Research Center for Drug Evaluation of major chronic diseases, Hengyang, China.
| | - Yingying Shen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
- Hunan Provincial Clinical Medical Research Center for Drug Evaluation of major chronic diseases, Hengyang, China.
| |
Collapse
|
10
|
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] [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
|
11
|
Westerberg C, Mestre Borras A, Ståhl S, Löfblom J. Affibody-based HER2 prodrug shows conditional cytotoxic effect on HER2-positive cancer cells. Biochem Biophys Res Commun 2025; 758:151660. [PMID: 40117970 DOI: 10.1016/j.bbrc.2025.151660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Therapeutic affinity proteins offer a targeted mode of action due to their typically high affinity and specificity for disease-associated molecules. In cancer therapy, such target molecules are often overexpressed receptors on tumor cells. However, their presence in healthy tissues can lead to on-target, off-tumor toxicity, necessitating strategies to enhance tumor selectivity. Here, we present an affibody-based prodrug concept that exploits tumor-associated proteases for selective activation. As proof of concept, we designed, produced, and characterized HER2-specific prodrug candidates, each incorporating a distinct protease substrate for selective activation by tumor-associated proteases. Their activation by corresponding proteases and subsequent HER2 binding were assessed. The most promising prodrug candidate was conjugated to the cytotoxic agent DM1 and evaluated for cytotoxicity in HER2-positive cancer cells. The results demonstrated potent, HER2-dependent cell killing, with markedly reduced cytotoxicity in the absence of prodrug activation. These findings support the feasibility of affibody-based prodrugs as a strategy to enhance tumor selectivity and minimize off-tumor toxicity in targeted cancer therapy.
Collapse
Affiliation(s)
- Cornelia Westerberg
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Anna Mestre Borras
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Stefan Ståhl
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
12
|
Zhao W, Feng X, Yang S, Yuan G, Huang M, Ding L, He Z, Wu J. Ophthalmotoxicity induced by antibody-drug conjugates: a pharmacovigilance study of the FDA adverse event reporting system (FAERS). Expert Opin Drug Saf 2025:1-11. [PMID: 40202446 DOI: 10.1080/14740338.2025.2491125] [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: 11/14/2024] [Revised: 03/07/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Antibody-drug conjugates (ADCs) have demonstrated remarkable therapeutic efficacy in refractory cancers, however, ophthalmotoxicity remains a serious concern. This study aimed to investigate the association between ADCs and ophthalmotoxicity. RESEARCH DESIGN AND METHODS A retrospective pharmacovigilance study was conducted utilizing data extracted from the U.S. Food and Drug Administration Adverse Events Reporting System (FAERS) from 2004 to 2023. Disproportionality analyses were performed using the reporting odds ratio (ROR) and information component (IC), with sensitivity analyses and subgroup evaluations by age and sex. RESULTS A total of 1992 cases of ophthalmotoxicity linked to ADCs were identified, with a median latency of 40 days. The correlation between ophthalmotoxicity and ADCs was higher than with other medications (IC = 0.67, 95% CI:0.64-0.70). Signal detection revealed 36 adverse events unreported in product labeling. Sensitivity analyses confirmed the robustness of our results on the association between ADCs and ocular toxicity, with higher reporting in females compared to males (OR = 1.25, 95% CI: 1.11-1.40). CONCLUSIONS ADCs had different profiles of ophthalmotoxicity. Our pharmacovigilance study suggested increased reporting of ophthalmotoxicity associated with ADCs.
Collapse
Affiliation(s)
- Wenxia Zhao
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Xin Feng
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Shan Yang
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Guosen Yuan
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Min Huang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - LinXiaoxiao Ding
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhichao He
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Junyan Wu
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| |
Collapse
|
13
|
Wang S, Wang X, Xia J, Mu Q. Identification of M1 macrophage infiltration-related genes for immunotherapy in Her2-positive breast cancer based on bioinformatics analysis and machine learning. Sci Rep 2025; 15:12525. [PMID: 40216945 PMCID: PMC11992169 DOI: 10.1038/s41598-025-96917-4] [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: 08/26/2024] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Over the past several decades, there has been a significant increase in the number of breast cancer patients. Among the four subtypes of breast cancer, Her2-positive breast cancer is one of the most aggressive breast cancers. In this study, we screened the differentially expressed genes from The Cancer Genome Atlas-Breast cancer database and analyzed the relationship between immune cell infiltration and differentially expressed genes using weighted gene co-expression network analysis. By constructing a module-trait relationships heatmap, the red module, which had the highest correlation value with M1 macrophages, was selected. Twenty hub genes were selected based on a protein-protein interaction network. Then, four overlapping M1 macrophage infiltration-related genes (M1 MIRGs), namely CCDC69, PPP1R16B, IL21R, and FOXP3, were obtained using five machine-learning algorithms. Subsequently, nomogram models were constructed to predict the incidence of Her2-positive breast cancer patients. The outer datasets and receiver operating characteristic curve analysis were used to validate the accuracy of the four M1 MIRGs and nomogram models. The average value of the area under the curve for the nomogram models was higher than 0.75 in both the training and testing sets. After that, survival analysis showed that higher expression of CCDC69, PPP1R16B, and IL21R were associated with overall survival of Her2-positive breast cancer patients. The expression of CCDC69 and PPP1R16B could lead to more benefits than the expression of IL21R and FOXP3 for immunotherapy. Lastly, we conducted immunohistochemistry staining to validate the aforementioned results. In conclusion, we found four M1 MIRGs that may be helpful for the diagnosis, prognosis, and immunotherapy of Her2-positive breast cancer.
Collapse
Affiliation(s)
- Sizhang Wang
- Qingdao Medical College of Qingdao University, Qingdao, 266042, Shandong, China
- Department of Breast surgery, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266042, Shandong, China
| | - Xiaoyan Wang
- General Practice Department, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266042, Shandong, China
| | - Jing Xia
- Department of Breast surgery, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266042, Shandong, China
| | - Qiang Mu
- Department of Breast surgery, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266042, Shandong, China.
| |
Collapse
|
14
|
Chazeau E, Pipier A, Wegner KD, Ghiringhelli F, Sancey L, Paul C, Goze C. NIR-II aza-BODIPY Platform for the Development of a Fluorescent Antibody Drug Conjugate. J Med Chem 2025; 68:7232-7242. [PMID: 40152348 DOI: 10.1021/acs.jmedchem.4c02777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Real-time imaging of antibody-drug conjugates (ADCs) offers valuable insights for assessing tumor targeting specificity, monitoring therapeutic efficacy, and detecting off-target accumulation that may cause adverse effects. To enable precise tracking, we developed a versatile fluorescent platform based on an NIR-II emitting aza-BODIPY dye, which can be site-specifically grafted onto an IgG1 antibody to generate well-defined fluorescent ADCs. As a proof of concept, we synthesized an HER2-targeting trastuzumab immunoconjugate bearing a NIR-II aza-BODIPY fluorophore. The cytotoxic monomethyl auristatin E (MMAE) payload was introduced in the final step, resulting in a trackable and homogeneous ADC suitable for both in vitro and in vivo investigations. The resulting Trastu-azaNIRII-MMAE selectively accumulated in HER2-positive subcutaneous tumors, significantly reducing the tumor growth. Using NIR-II optical imaging, a single injection of the NIR-II-ADC allowed for the detection of the conjugate over a period of more than one month, highlighting its potential for long-term tracking and therapeutic applications.
Collapse
Affiliation(s)
- Elisa Chazeau
- ICMUB, UMR 6302 CNRS, Université Bourgogne Europe, 9 av. A. Savary, BP 47870, 21078 Dijon, France
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France
- CTM, UMR1231 INSERM, Université de Bourgogne, 21000 Dijon, France
| | - Angélique Pipier
- ICMUB, UMR 6302 CNRS, Université Bourgogne Europe, 9 av. A. Savary, BP 47870, 21078 Dijon, France
| | - K David Wegner
- Division Biophotonics, Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| | - François Ghiringhelli
- CTM, UMR1231 INSERM, Université de Bourgogne, 21000 Dijon, France
- Plateforme de Transfert en Biologie Cancérologique, CGFL, 21000 Dijon, France
| | - Lucie Sancey
- Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000 Grenoble, France
| | - Catherine Paul
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France
- CTM, UMR1231 INSERM, Université de Bourgogne, 21000 Dijon, France
| | - Christine Goze
- ICMUB, UMR 6302 CNRS, Université Bourgogne Europe, 9 av. A. Savary, BP 47870, 21078 Dijon, France
| |
Collapse
|
15
|
Lee H, Ryu JM, Lee SK, Chae BJ, Yu J, Lee JE, Kim SW, Nam SJ, Cho YA, Cho EY. HER2 mRNA Score From Quantitative ERBB2 mRNA Expression of Oncotype Dx : Can It Replace the HER2 Immunohistochemistry? Am J Surg Pathol 2025:00000478-990000000-00499. [PMID: 40170597 DOI: 10.1097/pas.0000000000002396] [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: 04/03/2025]
Abstract
As novel human epidermal growth factor receptor-2 (HER2) and drug conjugates have proven to be effective treatments for both HER2-positive breast cancer (BC) and HER2-low BC, the need to develop more accurate methods to quantify HER2 status has increased. We compared the correlation between the HER2 mRNA score (HS), obtained using the Oncotype DX (ODX) test, and HER2 immunohistochemistry (IHC) to verify the accuracy of HER2 quantification and its correlation with clinicopathologic characteristics. We retrospectively collected ODX test data from 1524 estrogen-receptor positive, HER2-negative patients with BC. No significant differences in clinicopathologic characteristics, including ODX Recurrence Score (RS), were observed between HER2-0 and HER2-low BC. The median HS value of the HER2-low subgroup was significantly higher than that of the HER2-0 subgroup ( P <0.001) and increased significantly between the 4 subgroups classified by HER2 IHC ( P <0.001). The receiver operating characteristic curve showed acceptable utility in distinguishing HER2-0 from HER2-low (area under the curve=0.76) and HER2-null and HER2-ultralow subgroups (area under the curve=0.81), but the overlap between subgroups was also prominent. After defining 8.9 as a suboptimal cutoff mRNA score, multivariate analysis revealed that low Ki-67 (<20%) and RS (≤25) were statistically associated with higher HS (>8.9), whereas progesterone receptor negativity, high Ki-67, high nuclear grade, lower HS, and older age (>50 y) were statistically associated with high RS. Our study revealed that HS is significantly associated with HER2 IHC results and clinicopathologic parameters other than HER2 IHC.
Collapse
Affiliation(s)
- Hyunwoo Lee
- Department of Pathology and Translational Genomics
| | - Jai Min Ryu
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se Kyung Lee
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Byung Joo Chae
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jonghan Yu
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeong Eon Lee
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seok Won Kim
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seok Jin Nam
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoon Ah Cho
- Department of Pathology and Translational Genomics
| | - Eun Yoon Cho
- Department of Pathology and Translational Genomics
| |
Collapse
|
16
|
El Gazzah E, Parker S, Pierobon M. Multi-omic profiling in breast cancer: utility for advancing diagnostics and clinical care. Expert Rev Mol Diagn 2025:1-17. [PMID: 40193192 DOI: 10.1080/14737159.2025.2482639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 03/18/2025] [Indexed: 04/09/2025]
Abstract
INTRODUCTION Breast cancer remains a major global health challenge. While advances in precision oncology have contributed to improvements in patient outcomes and provided a deeper understanding of the biological mechanisms that drive the disease, historically, research and patients' allocation to treatment have heavily relied on single-omic approaches, analyzing individual molecular dimensions such as genomics, transcriptomics, or proteomics. While these have provided deep insights into breast cancer biology, they often fail to offer a complete understanding of the disease's complex molecular landscape. AREAS COVERED In this review, the authors explore the recent advancements in multi-omic research in the realm of breast cancer and use clinical data to show how multi-omic integration can offer a more holistic understanding of the molecular alterations and their functional consequences underlying breast cancer. EXPERT OPINION The overall developments in multi-omic research and AI are expected to complement precision diagnostics through potentially refining prognostic models, and treatment selection. Overcoming challenges such as cost, data complexity, and lack of standardization is crucial for unlocking the full potential of multi-omics and AI in breast cancer patient care to enable the advancement of personalized treatments and improve patient outcomes.
Collapse
Affiliation(s)
- Emna El Gazzah
- School of Systems Biology, George Mason University, Manassas, VA, USA
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Scott Parker
- School of Systems Biology, George Mason University, Manassas, VA, USA
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Mariaelena Pierobon
- School of Systems Biology, George Mason University, Manassas, VA, USA
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| |
Collapse
|
17
|
Lam C, Varghese D, Collins J, Nordstrom B, Murphy B, Mehta S, Faherty E. Treatment patterns and associated outcomes among patients with HER2+ metastatic breast cancer in the United States: an observational cohort study. Oncologist 2025; 30:oyae280. [PMID: 40192324 PMCID: PMC11973896 DOI: 10.1093/oncolo/oyae280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/10/2024] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND With treatment options for human epidermal growth factor receptor 2-positive (HER2+) metastatic breast cancer (mBC) expanding, updated assessments of contemporary treatment patterns and clinical outcomes are needed. This study aimed to conduct such an assessment using data from real-world oncology practices. MATERIALS AND METHODS Adult HER2+ mBC patients initiating first-line (1L) treatment from January 2013 to January 2021 (index date) were selected from the US Flatiron Health database and followed through January 2022. Patient characteristics and treatment patterns were summarized. Clinical outcomes were examined using Kaplan-Meier analyses. RESULTS Among 2074 HER2+ mBC patients with at least 1 line of therapy (LoT), median age was 61 years and 62.8% had known hormone receptor-positive disease. During a median follow-up of 26.0 months, 1159 (55.8%) had at least 2 LoTs, and 584 (28.2%) had 3 or more. The most common 1L regimens included docetaxel, trastuzumab, and pertuzumab (THP; 38.9%) followed by THP+ platinum agent (7.5%) and ado-trastuzumab emtansine (T-DM1) monotherapy (6.1%). By the end of follow up, 18.1% of patients remained on treatment, 20.2% died, and 5.8% discontinued without starting a new treatment. Median overall survival from 1L start was 40.3 months. CONCLUSIONS Approximately one-quarter of the patients died or discontinued 1L therapy without receiving further treatment. Overall survival from the start of 1L was just over 3 years. This highlights a need for more effective therapies in earlier LoTs that prolong the time to progression and provide longer clinical benefits.
Collapse
Affiliation(s)
- Clara Lam
- AstraZeneca Pharmaceuticals, LP, Gaithersburg, MD, United States
| | - Della Varghese
- AstraZeneca Pharmaceuticals, LP, Gaithersburg, MD, United States
| | | | | | | | - Sandhya Mehta
- Daiichi Sankyo, Inc., Basking Ridge, NJ, United States
| | | |
Collapse
|
18
|
Mortaja M, Adams SR, McKay RR, Gutkind JS, Advani SJ. Spatially precise chemo-radio-immunotherapy by antibody drug conjugate directed tumor radiosensitization to potentiate immunotherapies. NPJ Precis Oncol 2025; 9:97. [PMID: 40181161 PMCID: PMC11968929 DOI: 10.1038/s41698-025-00885-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
Concurrent chemo-radiotherapy is standard of care for locally advanced cancer patients. While radiotherapy and immuno-oncology have advanced precision oncology, chemotherapies in the chemo-radiotherapy paradigm remain non-targeted cytotoxins. Antibody drug conjugates offer an opportunity for targeted radiosensitization that stimulates immune responses while protecting normal tissues. Here, we discuss the rationale for combining antibody drug conjugates, radiotherapy and immunotherapies and opportunities for clinical translation to advance towards targeted chemo-radio-immunotherapy precision cancer care.
Collapse
Affiliation(s)
- Mahsa Mortaja
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Stephen R Adams
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Rana R McKay
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- UC San Diego, Moores Cancer Center, La Jolla, CA, 92093, USA
- Department of Urology, University of California San Diego, La Jolla, CA, 92093, USA
| | - J Silvio Gutkind
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
- UC San Diego, Moores Cancer Center, La Jolla, CA, 92093, USA
| | - Sunil J Advani
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, 92093, USA.
- UC San Diego, Moores Cancer Center, La Jolla, CA, 92093, USA.
| |
Collapse
|
19
|
Lázaro J, Joven T, Széliová D, Zanghellini J, Júlvez J. Multi-scale design and optimization of antibody production via flexible nets. Comput Struct Biotechnol J 2025; 27:1498-1510. [PMID: 40265159 PMCID: PMC12013398 DOI: 10.1016/j.csbj.2025.03.040] [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: 12/17/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Antibodies are therapeutic proteins with many applications in medicine, such as treating viral infections, different types of cancer, and common diseases such as psoriasis and multiple sclerosis. Chinese Hamster Ovary (CHO) cells are the most widely used cells for antibody production due to their well-established use and favorable features. However, the current design of antibody production systems often relies on a "trial and error" approach to manipulate CHO cells. This approach is time-consuming and costly, and can lead to suboptimal process performance. The use of mathematical models has the potential to greatly accelerate and improve the design and optimization of antibody production. Starting from a systematic and formal approach, the aim is to achieve an automatic design of the whole process that allows optimal productivity to be reached. To this end, we develop mathematical models and methods for the design and optimization of antibody manufacturing systems. The mathematical models are based on Flexible Nets (FNs), a modeling formalism that accommodates uncertain parameters and nonlinear dynamics. FNs enable the development of comprehensive models that encompass both the metabolic network of CHO cells and the dynamics of the bioreactor in which the cells are cultured. Thus, by integrating macroscopic variables (e.g. dilution rate, substrate concentration, cell density, etc.) with microscopic variables (such as intracellular metabolic fluxes), our model represents a multi-scale system and facilitates global optimization.
Collapse
Affiliation(s)
- Jorge Lázaro
- Department of Computer Science and Systems Engineering, University of Zaragoza, Zaragoza, Spain
| | - Teresa Joven
- Department of Computer Science and Systems Engineering, University of Zaragoza, Zaragoza, Spain
| | - Diana Széliová
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Jürgen Zanghellini
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Jorge Júlvez
- Department of Computer Science and Systems Engineering, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
20
|
Chiang ZC, Xu S, Zhao X, Liu M, Lin J, Chen Q. Generation and characterization of 7DC-DM1: a non-cleavable CD47-targeting antibody-drug conjugates with antitumor effects. Int J Biol Macromol 2025; 310:142844. [PMID: 40187444 DOI: 10.1016/j.ijbiomac.2025.142844] [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: 11/06/2024] [Revised: 03/30/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Colorectal cancer is the second leading cause of cancer-related deaths following lung cancer in recent years. Therefore, lung or colorectal cancer therapy is very important for reducing mortality. In this study, we developed and characterized CD47-specific antibody-drug conjugates, namely 7DC-DM1 ADCs, to evaluate their therapeutic effects on lung and colorectal cancer. Both 7DC2-DM1 and 7DC4-DM1 demonstrated good binding affinities of 0.56 nM and 0.49 nM, respectively, and exhibited significant cytotoxicity, though they displayed different penetration effects. These findings suggest that the binding complexes of 7DC2-DM1 and 7DC4-DM1 with CD47 receptors adopt different conformations, leading to variations in their cellular internalized efficiencies. Molecular docking simulations revealed that 7DC2 and 7DC4 bind to CD47 molecules in distinct orientations and epitopes, differing between conserved and non-conserved regions. Furthermore, treatments with 7DC2-DM1 and 7DC4-DM1 displayed notable differences in antitumor effects in murine syngeneic tumor models derived from the MC38 cell line in C57BL/6 mice. In the tumor model treated with 7DC4-DM1, immunofluorescence staining analysis revealed a large area of necrosis in the tumor stroma, accompanied by a significant infiltration of CD11b-expressing immune cells. In summary, these results indicate that 7DC4-DM1 holds promise as a therapeutic agent for colorectal cancer treatment.
Collapse
Affiliation(s)
- Zu-Chian Chiang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China; College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, China.
| | - Shan Xu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China
| | - Xiangqian Zhao
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China
| | - Min Liu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China.
| | - Jizhen Lin
- The Cancer Center, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China.
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China.
| |
Collapse
|
21
|
Inoue K, Nakamura Y, Caughey B, Zheng-Lin B, Ueno M, Furukawa M, Kawamoto Y, Itoh S, Umemoto K, Sudo K, Satoh T, Mizuno N, Kajiwara T, Fujisawa T, Bando H, Yoshino T, Strickler JH, Morizane C, Bekaii-Saab T, Ikeda M. Clinicomolecular Profile and Efficacy of Human Epidermal Growth Factor Receptor 2 (HER2)-Targeted Therapy for HER2-Amplified Advanced Biliary Tract Cancer. JCO Precis Oncol 2025; 9:e2400718. [PMID: 40209139 PMCID: PMC12005869 DOI: 10.1200/po-24-00718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/17/2024] [Accepted: 02/19/2025] [Indexed: 04/12/2025] Open
Abstract
PURPOSE This study aimed to investigate the clinicomolecular profiles and the efficacy of human epidermal growth factor receptor 2 (HER2)-targeted therapy in HER2-amplified biliary tract cancer (BTC). METHODS This study was an international collaboration that used combined data from the prospective SCRUM-Japan GOZILA and MONSTAR-SCREEN in Japan and retrospective reviews in the United States; patients with advanced BTC who had received systemic therapy were included. The clinicomolecular profiles were evaluated in an exploratory cohort, whereas the efficacy of HER2-targeted therapy was assessed in a biomarker-selected cohort. RESULTS Of the 439 patients in the exploratory cohort, 43 (10%) had HER2 amplification. The frequencies of coalterations were higher in patients with HER2 amplification versus patients without HER2 amplification including HER2 mutations (26% v 5%, P < .001), TP53 mutations (84% v 61%, P = .003), and BRAF amplification (9% v 2%, P = .030). There were no KRAS mutations identified in patients with HER2-amplified BTC. No significant difference in overall survival (OS) was observed between patients with and without HER2 amplification (median, 17.7 v 16.9 months; hazard ratio [HR], 0.95 [95% CI, 0.65 to 1.40]). Of the 60 patients with HER2-amplified BTC in the biomarker-selected cohort (43 from Japan and 17 from the United States), the OS was significantly longer in 29 patients who received HER2-targeted therapy than in those who did not receive HER2-targeted therapy (median, 24.3 v 12.1 months; HR, 0.39 [95% CI, 0.23 to 0.82]). Multivariate analysis identified HER2-targeted therapy as an independent prognostic factor for OS (HR, 0.29 [95% CI, 0.14 to 0.58]; P < .001). CONCLUSION HER2 amplification was found in 10% of advanced BTC and was not identified as an independent prognostic factor for OS. Patients with HER2-amplified BTC derive significant benefit from HER2-targeted therapy.
Collapse
Affiliation(s)
- Kanae Inoue
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Bennett Caughey
- Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA
| | - Binbin Zheng-Lin
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Masayuki Furukawa
- Department of Hepato-Biliary-Pancreatology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Yasuyuki Kawamoto
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kumiko Umemoto
- Department of Clinical Oncology, St Marianna University School of Medicine, Kawasaki, Japan
| | - Kentaro Sudo
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Taroh Satoh
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Osaka, Japan
| | - Nobumasa Mizuno
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Takeshi Kajiwara
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Takao Fujisawa
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Translational Research Support Office, Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | | | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | | | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
22
|
Hao C, Wang X, Shi Y, Tong Z, Li S, Liu X, Zhang L, Zhang J, Meng W, Zhang L. Combination Therapy of Pyrotinib and Metronomic Vinorelbine in HER2+ Advanced Breast Cancer after Trastuzumab Failure (PROVE): A Prospective Phase 2 Study. Cancer Res Treat 2025; 57:434-442. [PMID: 39118524 DOI: 10.4143/crt.2024.340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/08/2024] [Indexed: 08/10/2024] Open
Abstract
PURPOSE Approximately 50%-74% of patients with metastatic human epidermal growth factor receptor 2 (HER2)-positive breast cancer do not respond to trastuzumab, with 75% of treated patients experiencing disease progression within a year. The combination of pyrotinib and capecitabine has showed efficacy in these patients. This study evaluates the efficacy and safety of pyrotinib combined with metronomic vinorelbine for trastuzumab-pretreated HER2-positive advanced breast cancer patients. MATERIALS AND METHODS In this phase 2 trial, patients aged 18-75 years with HER2-positive advanced breast cancer who had previously failed trastuzumab treatment were enrolled to receive pyrotinib 400 mg daily in combination with vinorelbine 40mg thrice weekly. The primary endpoint was progression-free survival (PFS), while secondary endpoints included objective response rate (ORR), disease control rate (DCR), overall survival (OS), and safety. RESULTS From October 21, 2019, to January 21, 2022, 36 patients were enrolled and received at least one dose of study treatment. At the cutoff date, 20 experienced disease progression or death. With a median follow-up duration of 35 months, the median PFS was 13.5 months (95% confidence interval [CI], 8.3 to 18.5). With all patients evaluated, an ORR of 38.9% (95% CI, 23.1 to 56.5) and a DCR of 83.3% (95% CI, 67.2 to 93.6) were achieved. The median OS was not reached. Grade 3 adverse events (AEs) were observed in 17 patients, with diarrhea being the most common (27.8%), followed by vomiting (8.3%) and stomachache (5.6%). There were no grade 4/5 AEs. CONCLUSION Pyrotinib combined with metronomic vinorelbine showed promising efficacy and an acceptable safety profile in HER2-positive advanced breast cancer patients after trastuzumab failure.
Collapse
Affiliation(s)
- Chunfang Hao
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Department of Breast Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Xu Wang
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Yehui Shi
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Zhongsheng Tong
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Shufen Li
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Xiaodong Liu
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Lan Zhang
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Jie Zhang
- Department of Breast Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Wenjing Meng
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| | - Li Zhang
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
23
|
Bonotto M, De Pieri G, Esposto R, Lay L, Aprile G, Puglisi F, Minisini AM. Antibody-drug conjugates in elderly patients with breast cancer. Breast 2025; 80:104428. [PMID: 40020509 PMCID: PMC11919621 DOI: 10.1016/j.breast.2025.104428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/03/2025] Open
Abstract
Breast cancer remains a leading cause of cancer-related mortality worldwide, with elderly patients (aged >65 years) comprising a substantial portion of those affected. The treatment of breast cancer in this population is often complicated by frailty, comorbidities and polypharmacy. This review explores the application of antibody-drug conjugates (ADCs), such as trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd) and sacituzumab govitecan (SG), in treating breast cancer among elderly populations. The underrepresentation of older patients in clinical trials complicates efficacy and safety assessments in this group. Current evidence indicates that ADCs are both effective and tolerable in elderly patients, demonstrating improved progression-free survival (PFS) and overall survival (OS) alongside a manageable safety profile. Data from several trials like the EMILIA, TH3RESA and DestinyBreast studies demonstrate that T-DM1 and T-DXd maintained benefit in PFS and OS for HER2-positive breast cancer in older patients, despite a slight increase in adverse events. The ASCENT and TROPiCS-02 trials further confirm that SG provides significant improvements in PFS and OS in elderly patients at the cost of an increase in some toxicity. Emerging ADCs, including datopotamab deruxtecan and ARX-788, show promise but lack extensive geriatric-specific data. While the ADCs offer encouraging results in terms of efficacy and safety, with appropriate dose adjustments, further research is needed to optimize their use in elderly patients with breast cancer.
Collapse
Affiliation(s)
- Marta Bonotto
- Department of Oncology, Academic Hospital of Udine ASUFC, Udine, Italy.
| | - Giulia De Pieri
- Department of Oncology, Academic Hospital of Udine ASUFC, Udine, Italy; Department of Medicine, University of Udine, Udine, Italy
| | - Rocco Esposto
- Department of Oncology, Academic Hospital of Udine ASUFC, Udine, Italy; Department of Medicine, University of Udine, Udine, Italy
| | - Ludovica Lay
- Department of Oncology, Academic Hospital of Udine ASUFC, Udine, Italy; Department of Medicine, University of Udine, Udine, Italy
| | - Giuseppe Aprile
- Department of Oncology, Academic Hospital of Udine ASUFC, Udine, Italy
| | - Fabio Puglisi
- Department of Medicine, University of Udine, Udine, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | | |
Collapse
|
24
|
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
|
25
|
Géraud A, Gougis P, de Nonneville A, Beaufils M, Bertucci F, Billon E, Brisou G, Gravis G, Greillier L, Guerin M, Mezni E, Mitry E, Noel R, Pignon J, Sabatier R, Seguin L, Spano JP, Vicier C, Viret F, Goncalves A, Ciccolini J. Pharmacology and pharmacokinetics of antibody-drug conjugates, where do we stand? Cancer Treat Rev 2025; 135:102922. [PMID: 40157134 DOI: 10.1016/j.ctrv.2025.102922] [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: 01/24/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/01/2025]
Abstract
Antibody-drug conjugates (ADCs) are a rising therapeutic class in oncology and hematology, with eleven drugs approved by the US Food and Drug Administration as of January 2025. These "magic bullets" have a complex structure, including a monoclonal antibody, a linker, attachment sites, and a payload usually disrupting microtubules, targeting DNA, or inhibiting topoisomerase 1. By targeting specific tumor antigens, they are expected to be exquisitely effective in releasing "supertoxic" payloads inside tumor cells after intracellular trafficking. Additionally, they may exert a bystander effect, wherein the released payloads act on neighboring cells, amplifying their therapeutic impact regardless of target expression. ADCs have been game-changing drugs to treat tumors with once dismal prognoses or with previously considered unactionable targets, such as HER2-low or triple-negative breast cancer. To what extent there is room for personalized medicine to improve the toxicity/efficacy ratio remains unknown. However, there are inherent issues related to the complexity of the pharmacokinetics of ADCs and their assessments: efficacy or toxicity may be influenced by the clearance of the intact ADC, the circulating payload, or the payload-linker complex. Deciphering these multifaceted exposure-outcomes relationships for both efficacy and safety endpoints, is critical for advancing precision medicine and enabling personalized dosing strategies. To improve future developments and broaden their therapeutic scope, several strategies can be developed, including developing adequate combinations with other treatment classes (cytotoxic agents, immune-checkpoint inhibitors, oral molecular-targeted therapies). In this review, we will discuss the PK/PD aspects of ADCs and their dosing to improve their use in current and future indications.
Collapse
Affiliation(s)
- Arthur Géraud
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France; COMPO Team, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, Aix Marseille University, 13009 Marseille, France.
| | - Paul Gougis
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), 75013 Paris, France; Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Assistance Publique - Hôpitaux de Paris (AP-HP), Clinical Investigation Center (CIC-1901), Department of Pharmacology, Pitié-Salpêtrière Hospital, Paris, France; Residual Tumor & Response to Treatment Laboratory, RT2Lab, INSERM, U932 Immunity and Cancer, Institut Curie, France
| | - Alexandre de Nonneville
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Mathilde Beaufils
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - François Bertucci
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Emilien Billon
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Gabriel Brisou
- Department of Hematology, Institut Paoli-Calmettes, CRCM, Aix-Marseille University,13009 Marseille, France
| | - Gwenaelle Gravis
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Laurent Greillier
- COMPO Team, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, Aix Marseille University, 13009 Marseille, France; Department of Multidisciplinary Oncology and Therapeutic Innovations, Assistance Publique-Hôpitaux de Marseille (AP-HM), Aix Marseille University (AMU), 13015 Marseille, France
| | - Mathilde Guerin
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Essia Mezni
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Emmanuel Mitry
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Robin Noel
- Department of Hematology, Institut Paoli-Calmettes, CRCM, Aix-Marseille University,13009 Marseille, France
| | - Joséphine Pignon
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Renaud Sabatier
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Lorène Seguin
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Jean-Philippe Spano
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), 75013 Paris, France
| | - Cécile Vicier
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Frederic Viret
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Anthony Goncalves
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Joseph Ciccolini
- COMPO Team, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, Aix Marseille University, 13009 Marseille, France; Biogenopole, La timone University Hospital of Marseille, Assistance Publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| |
Collapse
|
26
|
Lipert BA, Siemens KN, Khan A, Airey R, Dam GH, Lu M, Flinterman M, Yong Q, Lee TW, Hunter FW, Jamieson SMF. CRISPR screens with trastuzumab emtansine in HER2-positive breast cancer cell lines reveal new insights into drug resistance. Breast Cancer Res 2025; 27:48. [PMID: 40165206 PMCID: PMC11959757 DOI: 10.1186/s13058-025-02000-1] [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: 08/17/2024] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate that is an effective therapy for HER2-positive breast cancer; however, its efficacy is limited by drug resistance. While multiple mechanisms of resistance have been proposed, these are not yet well understood. Greater understanding of T-DM1 sensitivity and resistance could provide new combination strategies to overcome resistance or predictive biomarkers to guide therapy. METHODS We have conducted CRISPR/Cas9 functional genomics modifier screens in HER2-positive breast cancer cell lines to allow for unbiased discovery of T-DM1 sensitivity and resistance genes. Whole-genome knockout screens were carried out in MDA-MB-361 and MDA-MB-453 cells treated with T-DM1 and its payload cytotoxin DM1. Hits were validated in secondary T-DM1 screens using a focused single-guide RNA (sgRNA) library and subsequently by individual gene knockout. RESULTS The whole-genome CRISPR screens with T-DM1 and DM1 identified 599 genes as potential modifiers of T-DM1 sensitivity and resistance. Of these, 17 genes were significantly enriched and 3 genes depleted at P < 0.001 in either or both MDA-MB-361 and MDA-MB-453 libraries in the secondary screens. Among the top hits, were known T-DM1 sensitivity genes ERBB2 and SLC46A3, in addition to negative regulators of mTOR complex 1: TSC1 and TSC2. MDA-MB-453 clones with knockout of TSC1 or partial knockout of TSC2 were more resistant to T-DM1 than wild type cells in competition growth assays and to T-DM1 and other HER2 targeting therapies (T-DXd, lapatinib and neratinib) in growth inhibition assays, and had increased internalisation of T-DM1 at 6 h. T-DM1 and the mTOR inhibitor everolimus demonstrated synergistic activity at inhibiting cell proliferation at multiple T-DM1 concentrations across four HER2-positive breast cancer cell lines. CONCLUSIONS Our CRISPR screening approach with T-DM1 in HER2-positive breast cancer cell lines identified genes not previously implicated in T-DM1 sensitivity or resistance, including TSC1 and TSC2. These genes may inform new strategies to enhance T-DM1 therapy in the clinic.
Collapse
Affiliation(s)
- Barbara A Lipert
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Kyla N Siemens
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Aziza Khan
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Rebecca Airey
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Gech Heng Dam
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Man Lu
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Marcella Flinterman
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Queenie Yong
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Tet Woo Lee
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Francis W Hunter
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Oncology Therapeutic Area, Johnson and Johnson Innovative Medicine, Spring House, PA, USA
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
27
|
Kang S, Kim SB. Toxicities and management strategies of emerging antibody-drug conjugates in breast cancer. Ther Adv Med Oncol 2025; 17:17588359251324889. [PMID: 40151551 PMCID: PMC11946287 DOI: 10.1177/17588359251324889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 02/14/2025] [Indexed: 03/29/2025] Open
Abstract
Antibody-drug conjugates (ADCs) offer a promising therapeutic approach for various cancers, enhancing the therapeutic window while mitigating systemic adverse effects on healthy tissues. ADCs have achieved remarkable clinical success, particularly in treating breast cancer, becoming a standard therapy across all subtypes, including hormone receptor-positive, human epidermal growth factor receptor 2-positive, and triple-negative breast cancer. Although designed to selectively target antigens via monoclonal antibodies, ADCs can exhibit toxicity in normal tissues, often due to off-target effects of their cytotoxic payloads. Understanding and managing these toxicities according to established guidelines are crucial for enhancing ADC clinical efficacy, minimizing adverse events, and ultimately improving patient outcomes. This review comprehensively examines the toxicities of ADCs employed in breast cancer treatment and explores their management strategies. Furthermore, we investigate novel ADCs beyond trastuzumab deruxtecan and sacituzumab govitecan, evaluating their potential efficacy and corresponding safety profiles.
Collapse
Affiliation(s)
- Sora Kang
- Division of Hemato-Oncology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| |
Collapse
|
28
|
Li N, Yang L, Zhao Z, Du T, Liang G, Li N, Tang J. Antibody-drug conjugates in breast cancer: current evidence and future directions. Exp Hematol Oncol 2025; 14:41. [PMID: 40114224 PMCID: PMC11924693 DOI: 10.1186/s40164-025-00632-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
Antibody-drug conjugates (ADCs) are a rapidly evolving class of antitumor drugs and have already revolutionized the treatment strategy of many hematologic and solid cancers. So far, trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd), sacituzumab govitecan (SG) and datopotamab deruxtecan (Dato-DXd) are the four ADCs that have been approved by US food and drug administration (FDA) in treatment of breast cancer, and SKB264 has been approved by Chinese national medical products administration (NMPA). Many ADCs for treatment of breast cancer are currently being tested in late-phase clinical trials, with several encouraging results achieved recently. However, major issues arise during the use of ADCs, including emergence of acquired resistance, occurrence of treated-related toxicities, and identification of biomarkers of response and resistance. ADCs are being increasingly tested in combination with other agents, and novel next-generation ADC development is progressing rapidly. A better understanding of the design and development of ADCs will promote ADC development for cancer treatment. In this review, we aim to provide a broad overview of the design and the recent advances of ADCs in breast cancer. We also propose several notable future directions of ADCs in treatment of breast cancer.
Collapse
Affiliation(s)
- Ning Li
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lu Yang
- Department of Radiotherapy, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
- Shantou University Medical College, Shantou University, Shantou, 515000, China
| | - Zixuan Zhao
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Tian Du
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Gehao Liang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Na Li
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Jun Tang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
29
|
Mascolo A, Dell'Anno I, Dondi L, Scavone C, Capuano A, Giordano M, Cristinziano A, Tarantino D, Pani M, Masini C, Donati C, Rossin E, Lagana G, Ballerio A, Bagaglini G, Bonanni G, Vergati A, Cavaliere A, Celenza R, D'Arpino A, Martini N, Piccinni C. Facts and figures of the compassionate use of onco-hematological drugs from 2016 to 2021: Results from the multicentric observational study Compass-O. J Oncol Pharm Pract 2025:10781552251327117. [PMID: 40080877 DOI: 10.1177/10781552251327117] [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: 03/15/2025]
Abstract
BackgroundCompassionate drug use (CDU) is a topic of debate in the scientific community characterized by a lack of information, especially in the onco-hematology area.AimThe Compass-O study aimed to provide updated data on CDU of onco-hematological drugs for the period 2016-2021.MethodCompass-O is a retrospective observational study with data obtained from 7 Italian Units for cytotoxic drug preparations (Unità Farmaci Antiblastici, UFA). Drugs and Therapeutic Indications (TI) were described separately and as drug-TI combinations, providing the mean Patients Per Year (mPPY) and the Years of Use (YoU).ResultsA total of 783 requests of CDU were retrieved, referring to 156.3 mPPY (max: 272 in 2021; min: 84 in 2018). The 52.2% of subjects was female, and the 63.2% aged >60 years. A total of 709 (90.5%) CDUs referred to solid tumors and 74 (9.5%) to liquid tumors. CDU referred to 93 drug-TI combinations. The most recurrent drug-TIs were nivolumab-lung carcinoma (mPPY: 36.5; YoU: 2) for solid tumors, and azacytidine-acute myeloid leukemia (6.0; 1) for liquid tumors. The most frequent TIs were lung carcinoma (45.7; 6) and breast cancer (37.0; 4) for solid tumors, and chronic lymphocytic leukemia (5.0; 2) and multiple myeloma (4.7; 6) for liquid tumors.ConclusionCompass-O provided an updated framework of the CDU in onco-hematology, showing its large-scale phenomenon and the relevance of UFA as a data source, underlining the need for a national observatory to monitor CDU.
Collapse
Affiliation(s)
- Annamaria Mascolo
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Napoli, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Napoli, Italy
- Department of Life Science, Health, and Health Professions, Link Campus University, Roma, Italy
| | - Irene Dell'Anno
- Fondazione ReS (Ricerca e Salute - Health and Research Foundation), Roma, Italia
| | - Leonardo Dondi
- Fondazione ReS (Ricerca e Salute - Health and Research Foundation), Roma, Italia
| | - Cristina Scavone
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Napoli, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Napoli, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Napoli, Italy
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Napoli, Italy
| | | | | | - Domenico Tarantino
- Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Roma, Italia
| | - Marcello Pani
- Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Roma, Italia
| | - Carla Masini
- Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori" - IRCCS, Meldola, Italia
| | - Caterina Donati
- Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori" - IRCCS, Meldola, Italia
| | - Elisabetta Rossin
- ASST Fatebenefratelli Sacco, Milano, Italia
- ASST Valle Olona, Busto Arsizio, Italia
| | | | | | | | | | | | | | - Rosanna Celenza
- A.O. di Perugia - Ospedale Santa Maria della Misericordia, Perugia, Italia
| | | | - Nello Martini
- Fondazione ReS (Ricerca e Salute - Health and Research Foundation), Roma, Italia
| | - Carlo Piccinni
- Fondazione ReS (Ricerca e Salute - Health and Research Foundation), Roma, Italia
| |
Collapse
|
30
|
Ryspayeva D, Seyhan AA, MacDonald WJ, Purcell C, Roady TJ, Ghandali M, Verovkina N, El-Deiry WS, Taylor MS, Graff SL. Signaling pathway dysregulation in breast cancer. Oncotarget 2025; 16:168-201. [PMID: 40080721 PMCID: PMC11906143 DOI: 10.18632/oncotarget.28701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
This article provides a comprehensive analysis of the signaling pathways implicated in breast cancer (BC), the most prevalent malignancy among women and a leading cause of cancer-related mortality globally. Special emphasis is placed on the structural dynamics of protein complexes that are integral to the regulation of these signaling cascades. Dysregulation of cellular signaling is a fundamental aspect of BC pathophysiology, with both upstream and downstream signaling cascade activation contributing to cellular process aberrations that not only drive tumor growth, but also contribute to resistance against current treatments. The review explores alterations within these pathways across different BC subtypes and highlights potential therapeutic strategies targeting these pathways. Additionally, the influence of specific mutations on therapeutic decision-making is examined, underscoring their relevance to particular BC subtypes. The article also discusses both approved therapeutic modalities and ongoing clinical trials targeting disrupted signaling pathways. However, further investigation is necessary to fully elucidate the underlying mechanisms and optimize personalized treatment approaches.
Collapse
Affiliation(s)
- Dinara Ryspayeva
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
| | - William J. MacDonald
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Connor Purcell
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Tyler J. Roady
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
| | - Maryam Ghandali
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Nataliia Verovkina
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
- Department of Medicine, Hematology/Oncology Division, Lifespan Health System and Brown University, RI 02903, USA
| | - Martin S. Taylor
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
- Brown Center on the Biology of Aging, Brown University, RI 02903, USA
| | - Stephanie L. Graff
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Department of Medicine, Hematology/Oncology Division, Lifespan Health System and Brown University, RI 02903, USA
| |
Collapse
|
31
|
Xie J, Yang Z, Li Z, Zhang T, Chen H, Chen X, Dai Z, Chen T, Hou J. Triple-positive breast cancer: navigating heterogeneity and advancing multimodal therapies for improving patient outcomes. Cancer Cell Int 2025; 25:77. [PMID: 40045297 PMCID: PMC11881339 DOI: 10.1186/s12935-025-03680-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/07/2025] [Indexed: 03/09/2025] Open
Abstract
Triple-positive breast cancer (TPBC), a unique subtype of luminal breast cancer, is characterized by concurrent positivity for estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Owing to the crosstalk between the ER and HER2 signaling pathways, the standard of care and drug resistance of this particular subtype are difficult challenges. Recent research and clinical trials have indicated a shift in the treatment paradigm for TPBC from single-target therapies to multi-pathway, multitarget strategies aiming to comprehensively modulate intricate signaling networks, thereby overcoming resistance and enhancing therapeutic outcomes. Among multiple strategies, triple-drug therapy has emerged as a promising treatment modality, demonstrating potential efficacy in patients with TPBC. Moving forward, there is a critical need to perform in-depth analyses of specific mechanisms of cancer pathogenesis and metastasis, decipher the complex interactions between different genes or proteins, and identify concrete molecular targets, thus paving the way for the development of tailored therapeutic strategies to combat TPBC effectively.
Collapse
Affiliation(s)
- Jie Xie
- GuiZhou University Medical College, Guiyang, 550025, Guizhou Province, China
| | - Zhihui Yang
- Zunyi Medical University, No.6 Xuefu West Road, Zunyi, 563006, Guizhou Province, China
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Zhuolin Li
- GuiZhou University Medical College, Guiyang, 550025, Guizhou Province, China
| | - Tianyu Zhang
- Urology Department, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China
| | - Huan Chen
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Xueru Chen
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Zehua Dai
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Tao Chen
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Jing Hou
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China.
| |
Collapse
|
32
|
Cai J, Wang W, Cong D, Bai Y, Zhang W. Development of treatment strategies for advanced HER2-positive gastric cancer: Insights from clinical trials. Crit Rev Oncol Hematol 2025; 207:104617. [PMID: 39805409 DOI: 10.1016/j.critrevonc.2025.104617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
HER2-positive gastric cancer (GC), a unique molecular subtype, has garnered significant interest in recent years. Here, we review clinical trial data on advanced HER2-positive GC from the past 15 years. Trastuzumab plus standard chemotherapy remain the first-line treatment. The initial survival benefits conferred by immune checkpoint inhibitors plus trastuzumab and standard chemotherapy are encouraging. The combination of ramucirumab and mono-chemotherapy, as well as the antibody conjugated drug trastuzumab deruxtecan, is the recommended second-line regimen. Treatment with immune checkpoint inhibitors plus ramucirumab and mono-chemotherapy shows promise. Despite the limited treatment options for third line and beyond, development of novel therapeutic strategies is expected. Although clinical cure of advanced HER2-positive GC is unlikely, current clinical studies offer valuable insight into regimens that prolong survival.
Collapse
Affiliation(s)
- Jing Cai
- Department of Pediatrics, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Wanning Wang
- Department of Nephrology, the First Hospital of Jilin University, Changchun 130021, China
| | - Dan Cong
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yuansong Bai
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Wenlong Zhang
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
33
|
Ueda Y, Kiyonaka S, Selfors LM, Inoue K, Harada H, Doura T, Onuma K, Uchiyama M, Kurogi R, Yamada Y, Sun JH, Sakaguchi R, Tado Y, Omatsu H, Suzuki H, Aoun M, Nakayama T, Kajimoto T, Yano T, Holmdahl R, Hamachi I, Inoue M, Mori Y, Takahashi N. Intratumour oxidative hotspots provide a niche for cancer cell dissemination. Nat Cell Biol 2025; 27:530-543. [PMID: 39984655 DOI: 10.1038/s41556-025-01617-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/10/2025] [Indexed: 02/23/2025]
Abstract
Intratumour heterogeneity represents the hierarchical integration of genetic, phenotypic and microenvironmental heterogeneity. Although single-cell sequencing has clarified genetic and phenotypic variability, the heterogeneity of nongenetic, microenvironmental factors remains elusive. Here, we developed T-AP1, a tumour-targeted probe tracking extracellular H2O2, which allows the visualization and characterization of tumour cells exposed to oxidative stress, a hallmark of cancer. T-AP1 identified actively budding intratumour regions as H2O2-rich microenvironments (H2O2 hotspots), which were primarily established by neutrophils. Mechanistically, tumour cells exposed to H2O2 underwent partial epithelial-mesenchymal transition through p38-MYC axis activation and migrated away from H2O2 hotspots. This escape mechanism was absent in normal epithelial cells but prevalent in most cancers except NRF2-hyperactivated tumours, which exhibited abrogated p38 responses and enhanced antioxidant programmes, thus revealing an intrinsic stress defence programme in cancers. Together, T-AP1 enabled the identification of H2O2 hotspots that provide a niche for cancer cell dissemination, offering insights into metastasis initiation.
Collapse
Affiliation(s)
- Yoshifumi Ueda
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Shigeki Kiyonaka
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan.
- Department of Biomolecular Engineering, Nagoya University, Nagoya, Japan.
- Research Institute for Quantum and Chemical Innovation, Nagoya University, Nagoya, Japan.
| | - Laura M Selfors
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Keisuke Inoue
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Tomohiro Doura
- Department of Biomolecular Engineering, Nagoya University, Nagoya, Japan
| | - Kunishige Onuma
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Makoto Uchiyama
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Ryuhei Kurogi
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Yuji Yamada
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Jiacheng H Sun
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Reiko Sakaguchi
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Yuki Tado
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Haruki Omatsu
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Harufumi Suzuki
- Department of Biomolecular Engineering, Nagoya University, Nagoya, Japan
| | - Mike Aoun
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | - Takahiro Nakayama
- Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Taketoshi Kajimoto
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | | | - Rikard Holmdahl
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan.
| | - Nobuaki Takahashi
- Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Kyoto, Japan.
- The Hakubi Center for Advanced Research, Kyoto University, Kyoto, Japan.
| |
Collapse
|
34
|
Pourjamal N, Le Joncour V, Vereb G, Honkamaki C, Isola J, Leyton JV, Laakkonen P, Joensuu H, Barok M. Disitamab vedotin in preclinical models of HER2-positive breast and gastric cancers resistant to trastuzumab emtansine and trastuzumab deruxtecan. Transl Oncol 2025; 53:102284. [PMID: 39837059 PMCID: PMC11788861 DOI: 10.1016/j.tranon.2025.102284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/27/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Most HER2-positive breast or gastric cancers eventually become resistant to the approved anti-HER2 antibody-drug conjugates (ADC) trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd). Disitamab vedotin (DV) is a novel anti-HER2 ADC that binds to a different epitope on HER2 compared to trastuzumab. We assessed the efficacy of DV in breast and gastric cancer cell lines and xenografts, including tumor models resistant to T-DM1 and T-DXd. Additionally, we investigated whether combining two anti-HER2 ADCs could enhance the efficacy of the individual ADCs. METHODS The efficacy of DV, T-DM1, and T-DXd, both as single agents and in combinations, was assessed using an AlamarBlue cell proliferation assay in HER2-positive breast and gastric cancer cell lines, including those resistant to T-DM1 and T-DXd. The efficacy of DV was evaluated also in breast and gastric cancer SCID mouse xenografts that had progressed on T-DM1 and/or T-DXd. ADC combinations were tested in breast and gastric cancer xenografts. RESULTS DV was effective in cell lines resistant to T-DM1 and/or T-DXd, and it inhibited the growth of breast and gastric cancer xenografts that had progressed on T-DM1 and/or T-DXd. The combinations of DV plus T-DM1 and DV plus T-DXd showed greater efficacy than the corresponding single agents in both breast and gastric cancer cell lines and xenografts. CONCLUSIONS DV was effective in treating breast and gastric cancer xenograft tumors resistant to T-DM1 and/or T-DXd. The combination of DV with T-DM1 or T-DXd demonstrated promising activity.
Collapse
Affiliation(s)
- Negar Pourjamal
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Laboratory of Molecular Oncology, Biomedicum, University of Helsinki, Helsinki, Finland
| | - Vadim Le Joncour
- Neuroscience Center, Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - György Vereb
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; HUN-REN-UD Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Cilla Honkamaki
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Laboratory of Molecular Oncology, Biomedicum, University of Helsinki, Helsinki, Finland
| | - Jorma Isola
- Laboratory of Cancer Biology, Medical Faculty, University of Tampere, Tampere, Finland
| | - Jeffrey V Leyton
- School of Pharmaceutical Sciences, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Pirjo Laakkonen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Laboratory Animal Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland; ICAN Digital Precision Medicine Flagship Program, University of Helsinki, Helsinki, Finland
| | - Heikki Joensuu
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Laboratory of Molecular Oncology, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Oncology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Mark Barok
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Laboratory of Molecular Oncology, Biomedicum, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
35
|
Markides DM, Hita AG, Merlin J, Reyes-Gibby C, Yeung SCJ. Antibody-Drug Conjugates: The Toxicities and Adverse Effects That Emergency Physicians Must Know. Ann Emerg Med 2025; 85:214-229. [PMID: 39641680 DOI: 10.1016/j.annemergmed.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 12/07/2024]
Abstract
Antibody-drug conjugates are novel antineoplastic agents whose use is expanding, both in terms of the number of drugs and the number of patients being treated. This article reviews the known toxicities and complications of antibody-drug conjugates that are currently approved for the treatment of cancer in the United States, with a focus on their emergency presentation and management. Similar to many other cancer therapies, most antibody-drug conjugates can cause diarrhea, nausea/vomiting, rash, peripheral neuropathy, and cytopenia, which are generally treated following standard-of-care. Interstitial lung disease, which may mimic pneumonia and cause respiratory failure and death, has been seen with trastuzumab deruxtecan and mirvetuximab soravtansine; emergency treatment of this condition includes oxygenation, ventilatory support, and corticosteroids. Inotuzumab ozogamicin and gemtuzumab ozogamicin are both associated with sinusoidal obstruction syndrome, a potentially fatal liver dysfunction that presents with weight gain, fluid overload, and jaundice. Abnormal liver function tests in patients who have been recently treated with these agents should be cautiously evaluated. Cardiac adverse events with antibody-drug conjugates are rare, but trastuzumab emtansine and trastuzumab deruxtecan may cause a decrease in cardiac contractility, and heart rate corrected QT interval prolongation is a rare effect of trastuzumab deruxtecan. Ocular adverse events, especially blurred vision, and keratopathy, are common with mirvetuximab soravtansine and tisotumab vedotin. Progressive multifocal leukoencephalopathy has been reported with brentuximab vedotin and polatuzumab vedotin. Tumor lysis syndrome may occur after treatment with gemtuzumab ozogamicin, polatuzumab vedotin, and brentuximab vedotin. Patients receiving enfortumab vedotin or brentuximab vedotin may develop hyperglycemia, sometimes presenting as diabetic ketoacidosis. Tisotumab vedotin and trastuzumab emtansine are associated with bleeding; although it is minor in most cases, severe bleeding and intracranial hemorrhage have occurred. Several antibody-drug conjugates can cause an anaphylactoid infusion-related reaction, which occurs most commonly during or soon after infusion but may be delayed up to 24 hours. Further research is needed to establish the real-world incidence of rare complications and how often patients with these complications present to the emergency department.
Collapse
Affiliation(s)
| | - Angel Guido Hita
- Department of Emergency Medicine, MD Anderson Cancer Center, Houston, TX
| | - Jeffrey Merlin
- Department of Emergency Medicine, MD Anderson Cancer Center, Houston, TX
| | - Cielto Reyes-Gibby
- Department of Emergency Medicine, MD Anderson Cancer Center, Houston, TX
| | - Sai-Ching J Yeung
- Department of Emergency Medicine, MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
36
|
Bouguerra Zina B, Rousseau F, Fauquier S, Sabatier R, Kfoury M. Practical clinical management of ocular adverse events related to Antibody-Drug Conjugates in gynaecological malignancies. Cancer Treat Rev 2025; 134:102867. [PMID: 39970828 DOI: 10.1016/j.ctrv.2024.102867] [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: 09/16/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 02/21/2025]
Abstract
BACKGROUND The advent of Antibody-Drug Conjugates (ADC) represents a significant advancement in targeted therapy for gynaecological malignancies. However, the ocular toxicities associated with ADCs, particularly Tisotumab Vedotin (TV) and Mirvetuximab Soravtansine (MIRV) necessitate effective mitigation in order to optimise patient care. METHODS This review synthesises findings from clinical trials to delineate the spectrum of ocular adverse events induced by ADCs. The analysis focuses on the incidence, onset, severity and reversibility of adverse events. It examines the underlying mechanisms of toxicity and provides management strategies based on study protocols. RESULTS Adverse events mainly impact the anterior ocular segment, resulting in conjunctivitis and keratopathy. They affect up to 56 % of patients treated with MIRV and 50 to 60 % of those receiving TV. Symptoms like blurred vision, dryness and pain hinder the patient's quality of life. Events are CTCAE grade 3 or higher in less than 10 % of cases. The median time to onset is 1.3 months. However, ocular toxicity may appear up to 10 months after treatment initiation, indicating a need for prolonged vigilance. Primary prophylaxis calls for local corticotherapy, lubricants and in some cases, vasoconstrictors. Despite the potential for severity, most cases are reversible with local treatment and transient dose reduction and/or delay. Close monitoring is crucial for early detection and subsequent management. CONCLUSIONS Clinicians ought to be cognizant of the potential ocular toxicity of ADCs. Proactive prophylaxis, patient education and a multidisciplinary approach involving ophthalmologists are paramount to minimising the impact of these AEs. Further research is essential to measure the real outcome of preventive strategies and balance their benefits with potential short and long-term risks.
Collapse
Affiliation(s)
- Bochra Bouguerra Zina
- Department of Medical Oncology, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France
| | - Frédérique Rousseau
- Department of Medical Oncology, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France
| | | | - Renaud Sabatier
- Department of Medical Oncology, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France; CRCM, Predictive Oncology Laboratory, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France
| | - Maria Kfoury
- Department of Medical Oncology, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France.
| |
Collapse
|
37
|
Okines AFC, Curigliano G, Mizuno N, Oh DY, Rorive A, Soliman H, Takahashi S, Bekaii-Saab T, Burkard ME, Chung KY, Debruyne PR, Fox JR, Gambardella V, Gil-Martin M, Hamilton EP, Monk BJ, Nakamura Y, Nguyen D, O'Malley DM, Olawaiye AB, Pothuri B, Reck M, Sudo K, Sunakawa Y, Van Marcke C, Yu EY, Ramos J, Tan S, Bieda M, Stinchcombe TE, Pohlmann PR. Tucatinib and trastuzumab in HER2-mutated metastatic breast cancer: a phase 2 basket trial. Nat Med 2025; 31:909-916. [PMID: 39825152 PMCID: PMC11922774 DOI: 10.1038/s41591-024-03462-0] [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: 04/24/2024] [Accepted: 12/11/2024] [Indexed: 01/20/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2, also known as ERBB2) signaling promotes cell growth and differentiation, and is overexpressed in several tumor types, including breast, gastric and colorectal cancer. HER2-targeted therapies have shown clinical activity against these tumor types, resulting in regulatory approvals. However, the efficacy of HER2 therapies in tumors with HER2 mutations has not been widely investigated. SGNTUC-019 is an open-label, phase 2 basket study evaluating tucatinib, a HER2-targeted tyrosine kinase inhibitor, in combination with trastuzumab in patients with HER2-altered solid tumors. The study included a cohort of 31 heavily pretreated female patients with HER2-mutated metastatic breast cancer who were also HER2 negative per local testing. Hormone receptor (HR)-positive patients also received fulvestrant. The overall response rate (primary endpoint) was 41.9% (90% confidence interval (CI): 26.9-58.2). Secondary endpoints of duration of response and progression-free survival were 12.6 months (90% CI: 4.7 to not estimable) and 9.5 months (90% CI: 5.4-13.8), respectively. No new safety signals were detected. Responses were observed across various HER2 mutations, including mutations in the tyrosine kinase and extracellular domains. The chemotherapy-free regimen of tucatinib and trastuzumab showed clinically meaningful antitumor activity with durable responses and favorable tolerability in heavily pretreated patients with HER2 mutations. These data support further investigation of HER2-targeted therapies in this patient population. ClinicalTrials.gov registration: NCT04579380 .
Collapse
Affiliation(s)
| | - Giuseppe Curigliano
- Istituto Europeo di Oncologia, IRCCS, Milan, Italy
- University of Milano, Milan, Italy
| | | | - Do-Youn Oh
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | - Andree Rorive
- CHU Sart Tilman Liège, University of Liège, Liège, Belgium
| | - Hatem Soliman
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | | | - Mark E Burkard
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Ki Y Chung
- Prisma Health Institute, Greenville, SC, USA
| | - Philip R Debruyne
- Kortrijk Cancer Centre, General Hospital AZ Groeninge, Kortrijk, Belgium
- Medical Technology Research Centre (MTRC), School of Life Sciences, Anglia Ruskin University, Cambridge, UK
- School of Nursing and Midwifery, University of Plymouth, Plymouth, UK
| | - Jenny R Fox
- Rocky Mountain Cancer Center, Boulder, CO, USA
| | | | - Marta Gil-Martin
- Institut Català d'Oncologia L'Hospitalet-IDIBELL, Hospitalet de Llobregat, Spain
| | | | - Bradley J Monk
- Florida Cancer Specialists and Research Institute, West Palm Beach, FL, USA
| | | | - Danny Nguyen
- City of Hope National Medical Center, Duarte, CA, USA
| | - David M O'Malley
- The Ohio State University and James Comprehensive Cancer Center, Columbus, OH, USA
| | | | - Bhavana Pothuri
- Laura & Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Martin Reck
- Department of Thoracic Oncology, Airway Research Center North, Germany Center for Lung Disease, Grosshansdorf, Germany
| | | | - Yu Sunakawa
- St. Marianna University Hospital, Kawasaki, Japan
| | | | - Evan Y Yu
- Fred Hutchinson Cancer Center/University of Washington, Seattle, WA, USA
| | | | | | | | | | - Paula R Pohlmann
- University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
38
|
Li X, Zhang X, Yin S, Nie J. Challenges and prospects in HER2-positive breast cancer-targeted therapy. Crit Rev Oncol Hematol 2025; 207:104624. [PMID: 39826885 DOI: 10.1016/j.critrevonc.2025.104624] [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: 07/09/2024] [Revised: 12/29/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025] Open
Abstract
Breast cancer remains the most prevalent malignancy among women globally and ranks as the leading cause of cancer-related mortality in this demographic. Approximately 13 %-15 % of all breast cancer cases are classified as HER2-positive, a subtype associated with a particularly unfavorable prognosis. A large number of patients with HER2-positive breast cancer continue to face disease progression after receiving standardized treatment. Given these challenges, a thorough exploration into the mechanisms underlying drug resistance in HER2-targeted therapy is imperative. This review focuses on the factors related to drug resistance in HER2-targeted therapy, including tumor heterogeneity, antibody-binding efficacy, variations in the tumor microenvironment, and abnormalities in signal activation and transmission. Additionally, corresponding strategies to counteract these resistance mechanisms are discussed, to advance therapeutic efficacy and clinical benefits in the management of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Xiyin Li
- Department of Breast Cancer, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, the Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan 650118, China.
| | - Xueying Zhang
- Department of Breast Cancer, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, the Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan 650118, China.
| | - Saige Yin
- Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650118, China.
| | - Jianyun Nie
- Department of Breast Cancer, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, the Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan 650118, China.
| |
Collapse
|
39
|
Lebow ES, Eichholz J, Zhang Z, Toumbacaris N, Imber B, Chen L, LaPlant Q, Yamada J, Pike LRG, Modi S, Seidman AD, Beal K, Moss NS, Yu Y. Local Therapy for Isolated Central Nervous System Progression Among Patients Receiving Antibody-Drug Conjugate Therapy. Adv Radiat Oncol 2025; 10:101714. [PMID: 40092156 PMCID: PMC11910671 DOI: 10.1016/j.adro.2025.101714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 01/03/2025] [Indexed: 03/19/2025] Open
Abstract
Purpose Antibody drug conjugates (ADCs) are an increasingly important class of therapeutics among patients with breast, lung, urothelial, and other malignancies. Guidelines recommend local therapy and continuation of current systemic therapy among patients with isolated brain relapse. We describe the clinical outcomes of this approach among patients receiving ADCs. Methods and Materials We queried our institutional database for patients receiving radiation therapy (RT) in the setting of isolated brain progression on ADCs with a plan to continue same-line therapy after radiation. Patients with ≤3 brain metastases at the time of recurrence were categorized as oligoprogressive. Study endpoints included overall survival, progression-free survival (PFS), and the cumulative incidence of next therapy from the start of local therapy. Results We identified 17 patients receiving ADC therapy with isolated brain progression treated with radiation (stereotactic radiosurgery [SRS]: n = 13, whole brain radiation: n = 4). All patients received concurrent ADC and RT. The median follow-up from local therapy was 29.5 months (95% CI, 21.4-not reached). The median overall survival was 19 months (95% CI, 16-not reached), and the median PFS was 8.1 months (range, 6.7-19 months). One lesion treated with SRS had local failure 21 months after treatment, and the 24-month cumulative incidence of local failure across the entire cohort was 1.6% (95% CI, 0.13%-7.7%). The 6-month cumulative incidence of radiation necrosis was 12% (95% CI, 1.8%-32%). The cumulative incidence of next therapy at 6 and 12 months was 47% (95% CI, 22%-69%) and 71% (95% CI, 41%-87%), respectively, and was significantly lower among patients with oligoprogressive brain recurrence. After SRS, 2 patients were without evidence of disease, discontinued systemic therapy, and were stable on observation at last follow-up. Conclusions To the best of our knowledge, this is the first clinical report of outcomes using the guideline-recommended approach of local therapy for isolated brain relapse among patients receiving ADCs. Local therapy may delay the need for next line systemic therapy, particularly among patients with oligoprogressive brain relapse.
Collapse
Affiliation(s)
- Emily S Lebow
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jordan Eichholz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zhigang Zhang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nicolas Toumbacaris
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brandon Imber
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Linda Chen
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Quincey LaPlant
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Josh Yamada
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Luke R G Pike
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shanu Modi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew D Seidman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kathryn Beal
- Department of Radiation Oncology, Cornell Medical Center, New York, New York
| | - Nelson S Moss
- Department of Neurosurgery and Brain Metastasis Center, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yao Yu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
40
|
Wei H, Zhang Y, Lu Y, Zou Y, Zhou L, Qin X, Jiang Q. Is ADC a rising star in solid tumor? An umbrella review of systematic reviews and meta-analyses. BMC Cancer 2025; 25:380. [PMID: 40021960 PMCID: PMC11871788 DOI: 10.1186/s12885-025-13726-8] [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: 01/17/2025] [Accepted: 02/12/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Antibody-drug conjugates (ADCs) combine the specificity of monoclonal antibodies with the potency of highly cytotoxic drugs and are known as panaceas, completely changing the treatment paradigm for solid tumors. Compared with other anti-cancer drugs, do they have better efficacy and lower toxicity risks? It is necessary to summarize and analyze the published clinical research data in this area to provide additional evidence-based evidence for clinical practice. OBJECTIVE To comprehensively assess and overview the efficacy and safety of antibody-drug conjugates for the treatment of solid tumors. DESIGN An umbrella review of systematic reviews and meta-analyses. METHODS Systematic search of eight electronic databases and one registration platform including Embase, PubMed, Cochrane Database of Systematic Reviews (CDSR), Web of Science (WoS), China National Knowledge Infrastructure (CNKI), Chinese BioMedical Literature Database (CBM), Wan Fang Data, China Science and Technology Journal Database (VIP) and international prospective register of systematic reviews (PROSPERO) on Aug 1, 2024, to identify relevant systematic reviews or meta-analyses. Three authors completed research screening and data extraction independently. AMSTAR 2 was used to evaluate the methodological quality of the included studies and the Grading of Recommendations Assessment, Development and Evaluation Working Group (GRADE) was performed to evaluate the quality of the evidence. We examined progression-free survival (PFS), overall survival (OS), objective response rate (ORR) as efficacy endpoints, and the incidence of adverse events (AEs) as safety profiles. RESULTS A total of 16 eligible publications, including 32 clinical studies, were included in the umbrella review. The methodological quality of the included study was poor, with 2 articles of moderate-quality (12.5%), 5 articles of low quality (31.25%), and 9 articles of critically low quality (56.25%). Only one third of the evidence was of high quality. Within the included studies, breast cancer accounted for four-fifths, 2 studies were gastric cancer, and 1 study was a solid tumor. The overall results showed that ADCs significantly increased PFS and OS in patients with solid tumors, and the risk of toxicity was within an acceptable range. ado-Trastuzumab emtansine (T-DM1) and Trastuzumab deruxtecan (T-Dxd) treatment of human epidermal growth factor receptor 2(HER2) low/positive advanced metastatic breast cancer significantly prolonged PFS and OS, but the ORR showed a significant advantage. Compared with the chemotherapy group, T-Dxd significantly prolonged OS and PFS in gastric cancer patients, while T-DM1 did not. In other cancer types (ovarian cancer, renal cell carcinoma, and malignant pleural mesothelioma), ADCs tended to extend overall survival or progression-free survival compared with controls, but the difference was not statistically significant. CONCLUSIONS Based on the available evidence, in breast cancer, ADCs were proved to with significant improvements in prolonging survival time and demonstrates a tolerable safety profile. Meanwhile, ADCs were proved to have enormous potential for the treatment of solid tumors. However, well-designed, multi-center RCTs need to further identify its potential in various solid tumors. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD 42,024,564,517.
Collapse
Affiliation(s)
- Hua Wei
- Department of Pharmacy, Chengdu Second People's Hospital, Chengdu, Sichuan, China
| | - Yongjun Zhang
- Department of Anesthesiology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, 610213, China
| | - Yun Lu
- Department of Pharmacy, Chengdu Second People's Hospital, Chengdu, Sichuan, China
| | - Ya Zou
- Department of Pharmacy, Chengdu Second People's Hospital, Chengdu, Sichuan, China
| | - Lu Zhou
- Department of Pharmacy, Chengdu Second People's Hospital, Chengdu, Sichuan, China
| | - Xiaoli Qin
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, 610031, Sichuan, P.R. China
| | - Qian Jiang
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, 610041, Sichuan, P.R. China.
| |
Collapse
|
41
|
Zhou Y, Gong J, Deng X, Shen L, Ge A, Fan H, Ling J, Wu S, Liu L. A comprehensive exploration of adverse reactions to lapatinib: a disproportionate analysis based on the FAERS database. Expert Opin Drug Saf 2025:1-10. [PMID: 39985750 DOI: 10.1080/14740338.2025.2471515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Lapatinib, an FDA-approved tyrosine kinase inhibitor, treats HER2+ advanced/metastatic breast cancer. This study comprehensively analyzed its adverse reaction profile using FDA Adverse Event Reporting System (FAERS) to guide clinical use. RESEARCH DESIGN AND METHODS Adverse event (AE) reports for lapatinib from the second quarter of 2007 to the second quarter of 2024 in FAERS were analyzed using Reporting Odds Ratio (ROR), Proportional Reporting Ratio (PRR), Multi-item Gamma Poisson Shrinkage (MGPS) and Bayesian Confidence Propagation Neural Network (BCPNN) to identify AE signals. RESULTS Among 8300 AE reports, females (91.47%) and ages 40-59.9 (33.71%) were predominant. 20 system organ classifications (SOCs) were affected, with gastrointestinal disorders (ROR = 3.46) and skin disorders (ROR = 2.47) most significant. Based on the PT level, a total of 111 PTs were analyzed that met the four algorithms, including typical AEs such as diarrhea (n = 3410), vomiting (n = 856), and rash (n = 856), as well as some rare AEs that were not prompted by the drug inserts, such as neutropenia (n = 252), pericardial effusion (n = 43), lymphedema (n = 20). The majority of lapatinib-associated AEs had onset within 30 days (51%). CONCLUSIONS Lapatinib has a generally favorable safety profile, but gastrointestinal toxicity and dermatotoxicity require close monitoring to prevent serious AEs.
Collapse
Affiliation(s)
- Yao Zhou
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jie Gong
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xianguang Deng
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lele Shen
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Anqi Ge
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hongqiao Fan
- Department of Aesthetic Plastic Surgery, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jie Ling
- Hunan Academy of Chinese Medicine, Changsha, Hunan, China
| | - Shiting Wu
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lifang Liu
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
42
|
Gu H, Zhu T, Ding J, Yang Z, Qi S, Guo G. Real-World Analysis of the Efficacy and Adverse Events of T-DM1 in Chinese Patients With HER2-Positive Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:201-210. [PMID: 40008213 PMCID: PMC11853105 DOI: 10.2147/bctt.s503150] [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: 11/07/2024] [Accepted: 01/25/2025] [Indexed: 02/27/2025]
Abstract
Purpose This study efforts to explore the association of adverse events (AEs) with efficacy in HER2-positive breast cancer patients treated with TDM1. Methods and Materials This retrospective study included women diagnosed with HER2+ BC treated with TDM1 from January 2012 to December 2023. Event-free survival (EFS) was the endpoint. Tumour response was assessed by disease control rate (DCR) and objective response rate (ORR). The chi-squared test, analysis of variance (ANOVA), Cox proportional hazards regression and Kaplan-Meier survival analysis was employed to evaluate the association of AEs with tumour efficacy. Results A total of 48 women with a median age of 52 years (median follow-up 8.4 months) were included in the study. Among them, 33 patients (68.8%) experienced adverse events, including platelet depletion and liver function abnormalities, 3 patients (6.3%) discontinued TDM1 due to severe platelet depletion. The overall objective response rate (ORR) was 25.0% and the disease control rate (DCR) was 43.8%. Using the Chi-squared test, we found a statistically significant difference in ORR and DCR between patients who developed a platelet reduction and those who did not. DCR was significantly higher in patients with liver dysfunction than in those without. ANOVA showed that exposure to hepatic dysfunction and platelet reduction, lines of therapy, and treatment course were associated with EFS. In the Kaplan-Meier survival analysis, both liver dysfunction and platelet reduction were correlated with significantly longer EFS (p=0.033 and p=0.038, respectively). Conclusion This retrospective study demonstrated that AEs were associated with tumour efficacy in patients with HER2+ BC treated with TDM1.
Collapse
Affiliation(s)
- Huayan Gu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Teng Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - JiaLing Ding
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Zhi Yang
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Shuangyi Qi
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Guilong Guo
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| |
Collapse
|
43
|
Xiong X, Zheng LW, Ding Y, Chen YF, Cai YW, Wang LP, Huang L, Liu CC, Shao ZM, Yu KD. Breast cancer: pathogenesis and treatments. Signal Transduct Target Ther 2025; 10:49. [PMID: 39966355 PMCID: PMC11836418 DOI: 10.1038/s41392-024-02108-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/27/2024] [Accepted: 12/08/2024] [Indexed: 02/20/2025] Open
Abstract
Breast cancer, characterized by unique epidemiological patterns and significant heterogeneity, remains one of the leading causes of malignancy-related deaths in women. The increasingly nuanced molecular subtypes of breast cancer have enhanced the comprehension and precision treatment of this disease. The mechanisms of tumorigenesis and progression of breast cancer have been central to scientific research, with investigations spanning various perspectives such as tumor stemness, intra-tumoral microbiota, and circadian rhythms. Technological advancements, particularly those integrated with artificial intelligence, have significantly improved the accuracy of breast cancer detection and diagnosis. The emergence of novel therapeutic concepts and drugs represents a paradigm shift towards personalized medicine. Evidence suggests that optimal diagnosis and treatment models tailored to individual patient risk and expected subtypes are crucial, supporting the era of precision oncology for breast cancer. Despite the rapid advancements in oncology and the increasing emphasis on the clinical precision treatment of breast cancer, a comprehensive update and summary of the panoramic knowledge related to this disease are needed. In this review, we provide a thorough overview of the global status of breast cancer, including its epidemiology, risk factors, pathophysiology, and molecular subtyping. Additionally, we elaborate on the latest research into mechanisms contributing to breast cancer progression, emerging treatment strategies, and long-term patient management. This review offers valuable insights into the latest advancements in Breast Cancer Research, thereby facilitating future progress in both basic research and clinical application.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Le-Wei Zheng
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu Ding
- Department of Breast and Thyroid, Guiyang Maternal and Child Health Care Hospital & Guiyang Children's Hospital, Guiyang, P. R. China
- Department of Clinical Medicine, Guizhou Medical University, Guiyang, P. R. China
| | - Yu-Fei Chen
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu-Wen Cai
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Lei-Ping Wang
- Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Liang Huang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China.
| |
Collapse
|
44
|
Lamtha T, Jongkon N, Lertvanithphol T, Horprathum M, Seetaha S, Choowongkomon K. Cannabinoids as Promising Inhibitors of HER2-Tyrosine Kinase: A Novel Strategy for Targeting HER2-Positive Ovarian Cancer. ACS OMEGA 2025; 10:6191-6200. [PMID: 39989803 PMCID: PMC11840771 DOI: 10.1021/acsomega.4c11108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/17/2025] [Accepted: 01/30/2025] [Indexed: 02/25/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a transmembrane receptor within the ErbB family that plays a pivotal role in the progression of various aggressive cancers. HER2-positive tumors often develop resistance to standard therapies, necessitating the exploration of innovative treatment options. Cannabinoids, bioactive compounds from Cannabis sativa such as cannabidiol (CBD), cannabigerol (CBG), and cannabinol (CBN), have gained attention for their potential anticancer properties. This study evaluates the efficacy of CBD, CBG, and CBN in targeting HER2-positive ovarian cancer through kinase inhibition assays, surface plasmon resonance (SPR), molecular docking, and cell viability assessments. SPR analysis revealed that cannabinoids bind strongly to HER2-tyrosine kinase (HER2-TK), with CBD showing the highest affinity (K D = 6.16 μM), significantly better than afatinib (K D = 26.30 μM), and CBG demonstrating moderate affinity (K D = 17.07 μM). In kinase inhibition assays, CBG was the most potent inhibitor (IC50 = 24.7 nM), followed by CBD (IC50 = 38 nM), suggesting their ability to disrupt HER2-mediated signaling pathways. Molecular docking studies highlighted critical interactions between cannabinoids and essential HER2 residues (Leu796, Thr862, Asp863). In cell viability assays, CBD and CBG effectively inhibited the growth of HER2-positive SKOV3 cells (IC50 = 13.8 μM and 16.6 μM, respectively), comparable to traditional tyrosine kinase inhibitors. These findings underscore the therapeutic potential of cannabinoids, particularly CBD and CBG, as alternative or adjunct therapies for HER2-positive cancers, with the promise of mitigating resistance and adverse effects associated with existing treatments.
Collapse
Affiliation(s)
- Thomanai Lamtha
- Laboratory
of Protein Engineering and Bioinformatics (PROTEP), Department of
Biochemistry, Faculty of Science, Kasetsart
University, Bangkok 10900, Thailand
- Department
of Clinical Chemistry, Faculty of Medical Technology, Nation University, Lampang 52000, Thailand
| | - Nathjanan Jongkon
- Department
of Social and Applied Science, College of Industrial Technology, King Mongkut’s University of Technology North
Bangkok, Bangkok 10800, Thailand
| | - Tossaporn Lertvanithphol
- The
National Electronics and Computer Technology Center (NECTEC), National
Science and Technology Development Agency (NSTDA), Thailand Science Park, Pathum
Thani 12120, Thailand
| | - Mati Horprathum
- The
National Electronics and Computer Technology Center (NECTEC), National
Science and Technology Development Agency (NSTDA), Thailand Science Park, Pathum
Thani 12120, Thailand
| | - Supaphorn Seetaha
- Laboratory
of Protein Engineering and Bioinformatics (PROTEP), Department of
Biochemistry, Faculty of Science, Kasetsart
University, Bangkok 10900, Thailand
| | - Kiattawee Choowongkomon
- Laboratory
of Protein Engineering and Bioinformatics (PROTEP), Department of
Biochemistry, Faculty of Science, Kasetsart
University, Bangkok 10900, Thailand
| |
Collapse
|
45
|
Hu X, Zhang Q, Wang L, Zhang J, Ouyang Q, Wang X, Li W, Xie W, Tong Z, Wang S, Xu F, Sun T, Liu W, Chen Z, Wu J, Wang Y, Wang H, Yan M, Wang X, Wang J, Cao F, Du Y, Zhang Y, Chen L, Lu P, Sun S, Zhang R, Zang A, Nie X, Lei Y. ACE-Breast-02: a randomized phase III trial of ARX788 versus lapatinib plus capecitabine for HER2-positive advanced breast cancer. Signal Transduct Target Ther 2025; 10:56. [PMID: 39956849 PMCID: PMC11830773 DOI: 10.1038/s41392-025-02149-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 01/06/2025] [Accepted: 01/16/2025] [Indexed: 02/18/2025] Open
Abstract
This phase III trial aimed to compare ARX788, a site-specific, construct-homogeneous antibody-drug conjugate, with lapatinib plus capecitabine in patients with human epidermal growth factor receptor 2 (HER2)-positive advanced breast cancer (ABC) who had progressed on one line of trastuzumab based regimen. Eligible patients were randomized (1:1) to receive ARX788 (1.5 mg/kg, IV, Q3W) or lapatinib plus capecitabine (LC: lapatinib 1250 mg QD; capecitabine 1000 mg/m2 BID, days 1-14, Q3W) and stratified by prior chemotherapy lines (0-1 versus >1) and visceral metastasis (yes versus no). The primary outcome was progression-free survival (PFS) assessed by a blinded independent central review (BICR). A total of 441 patients were randomly assigned to receive either ARX788 (n = 221) or LC (n = 220). The median PFS was 11.3 (95% confidence interval [CI], 8.4-13.8) months with ARX788 compared with 8.2 (95% CI, 6.9-8.7) months with LC, as per BICR (hazard ratio [HR] 0.64, p = 0.0006). Frequencies of treatment-related adverse events (TRAEs) of any grade were 98.6% and 99.1% for ARX788 and LC, respectively. Grade ≥3 TRAEs were 41.4% and 40.0%, respectively, the most common adverse events were blurred vision (12.3%), dry eye (9.1%), keratopathy (5.9%), and interstitial lung disease (ILD, 5.9%) with ARX788; hand-foot syndrome (18.1%) and hypokalemia (5.1%) with LC; all the hematological and gastrointestinal events of grade ≥3 with ARX788 were less than 3%. Six treatment-related deaths occurred, with three cases possibly related to ILD. ARX788 significantly improved PFS compared with LC in patients with HER2-positive ABC with a distinct toxicity profile, supporting it as a potential treatment option.
Collapse
Affiliation(s)
- Xichun Hu
- Fudan University Cancer Hospital, Shanghai, 200032, China.
| | - Qingyuan Zhang
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Leiping Wang
- Fudan University Cancer Hospital, Shanghai, 200032, China
| | - Jian Zhang
- Fudan University Cancer Hospital, Shanghai, 200032, China
| | | | - Xiaojia Wang
- Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Wei Li
- The First Hospital of Jilin University, Changchun, 130031, China
| | - Weimin Xie
- Guangxi Medical University Affiliated Tumor Hospital, Nanning, 530012, China
| | - Zhongsheng Tong
- Tianjin Medical University Cancer Institute & Hospital, Tianjin, 300060, China
| | - Shusen Wang
- Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Faliang Xu
- Chongqing University Cancer Hospital, Chongqing, China
| | - Tao Sun
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110122, China
| | - Wei Liu
- The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Zhendong Chen
- The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinsheng Wu
- The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Ying Wang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | | | - Min Yan
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xinshuai Wang
- The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | | | - Feilin Cao
- Taizhou Hospital, Wenzhou Medical University, Taizhou, China
| | - Yingying Du
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | | | - Lilin Chen
- First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Ping Lu
- The First Affiliated Hospital of Xinxiang Medical College, Xinxiang, China
| | | | | | - Aimin Zang
- Affiliated Hospital of Hebei University, Baoding, China
| | - Xiuqing Nie
- NovoCodex Biopharmaceuticals, Shaoxing, China
| | - Yuan Lei
- NovoCodex Biopharmaceuticals, Shaoxing, China
| |
Collapse
|
46
|
Ressnerova A, Heger Z, Pumera M. Translational nanorobotics breaking through biological membranes. Chem Soc Rev 2025; 54:1924-1956. [PMID: 39807638 DOI: 10.1039/d4cs00483c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
In the dynamic realm of translational nanorobotics, the endeavor to develop nanorobots carrying therapeutics in rational in vivo applications necessitates a profound understanding of the biological landscape of the human body and its complexity. Within this landscape, biological membranes stand as critical barriers to the successful delivery of therapeutic cargo to the target site. Their crossing is not only a challenge for nanorobotics but also a pivotal criterion for the clinical success of therapeutic-carrying nanorobots. Nevertheless, despite their urgency, strategies for membrane crossing in translational nanorobotics remain relatively underrepresented in the scientific literature, signaling an opportunity for further research and innovation. This review focuses on nanorobots with various propulsion mechanisms from chemical and physical to hybrid mechanisms, and it identifies and describes four essential biological membranes that represent the barriers needed to be crossed in the therapeutic journey of nanorobots in in vivo applications. First is the entry point into the blood stream, which is the skin or mucosa or intravenous injection; next is the exit from the bloodstream across the endothelium to the target site; further is the entry to the cell through the plasma membrane and, finally, the escape from the lysosome, which otherwise destroys the cargo. The review also discusses design challenges inherent in translating nanorobot technologies to real-world applications and provides a critical overview of documented membrane crossings. The aim is to underscore the need for further interdisciplinary collaborations between chemists, materials scientists and chemical biologists in this vital domain of translational nanorobotics that has the potential to revolutionize the field of precision medicine.
Collapse
Affiliation(s)
- Alzbeta Ressnerova
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00, Brno, Czech Republic.
- Research Group for Molecular Biology and Nanomedicine, Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic
| | - Zbynek Heger
- Research Group for Molecular Biology and Nanomedicine, Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic
- Center of Advanced Innovation Technologies, Faculty of Materials Science and Technology, VSB - Technical University of Ostrava, 17. Listopadu 2172/15, 70800 Ostrava, Czech Republic
| | - Martin Pumera
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00, Brno, Czech Republic.
- Advanced Nanorobots & Multiscale Robotics Laboratory, Faculty of Electrical Engineering and Computer Science, VSB - Technical University of Ostrava, 17. listopadu 2172/15, 70800 Ostrava, Czech Republic
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
- Department of Medical Research, China Medical University Hospital, China Medical University, No. 91 Hsueh-Shih Road, Taichung, Taiwan
| |
Collapse
|
47
|
Shi Z, Lu Y, Zhao Q, Wang Y, Qiu P. Antibody-drug conjugates in breast cancer: advances and prospects. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0486. [PMID: 39960255 PMCID: PMC11899594 DOI: 10.20892/j.issn.2095-3941.2024.0486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/17/2025] [Indexed: 03/14/2025] Open
Affiliation(s)
- Zhiqiang Shi
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300202, China
| | - Yongjin Lu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
- Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan 271016, China
| | - Qiuchen Zhao
- Cancer Research UK Cambridge Centre PhD Programme, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Yongsheng Wang
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Pengfei Qiu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| |
Collapse
|
48
|
Pougoue Ketchemen J, Njotu FN, Babeker H, Monzer A, Nwangele E, Tikum AF, Henning N, Hassani N, Frye S, Perron R, Byrne C, Didychuk C, Qi Q, Bannister L, Doroudi A, Fonge H. Complete Remissions of HER2-Positive Trastuzumab-Resistant Xenografts Using a Potent [225Ac]Ac-Labeled Anti-HER2 Antibody-Drug Radioconjugate. Clin Cancer Res 2025; 31:685-696. [PMID: 39670857 DOI: 10.1158/1078-0432.ccr-24-1779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/09/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024]
Abstract
PURPOSE There is overwhelming interest to use actinium-225 ([225Ac]Ac) to develop targeted α therapies. Antibody-drug conjugates (ADC) are highly cytotoxic. Combining [225Ac]Ac with an ADC to develop an antibody-drug radioconjugate [225Ac]Ac-macropa-trastuzumab(T)-PEG6-emtansine (DM1), is expected to be more effective than its ADC (T-PEG6-DM1) against breast cancer. EXPERIMENTAL DESIGN [89Zr]Zr-p-isothiocyanatobenzyl desferrioxamine (DFO)-T-PEG6-DM1 (imaging) and [225Ac]Ac-macropa-T-PEG6-DM1 (radiotherapy) were developed. Biodistribution and safety evaluations of [225Ac]Ac-macropa-T-PEG6-DM1 were carried out in non-tumor-bearing BALB/c mice. MicroPET imaging and biodistribution were done using [89Zr]Zr-DFO-T-PEG6-DM1, and radiotherapy using [225Ac]Ac-macropa-T-PEG6-DM1 was carried out in athymic BALB/c nude mice bearing trastuzumab-resistant HCC1954 and trastuzumab-DM1 (T-DM1)/trastuzumab-resistant JIMT-1 tumor-bearing mice. RESULTS After 7 days of incubation at 37°C, [225Ac]Ac-macropa-T-PEG6-DM1 was stable in both human serum (89.2% ± 0.9%) and PBS (82.8% ± 0.4%). T-PEG6-DM1 (8 mg/kg) and [225Ac]Ac-macropa-T-PEG6-DM1 (3 × 18 kBq) administered separately in non-tumor-bearing mice 10 days apart were well tolerated biochemically and hematologically. Imaging and biodistribution showed high tumor uptake of [89Zr]Zr-DFO-T-PEG6-DM1 in tumor-bearing mice at 120 hours after injection: 38.1% ± 2.8% IA/g (HCC1954) and 14.6% ± 1% IA/g (JIMT-1). In HCC1954 tumor-bearing mice, all treatment groups had complete remission (8/8), indicative of the responsiveness of the xenograft to T-DM1-based treatments, whereas for JIMT-1 xenografts (having 1/8 complete remission) at 23 days after treatment, tumor volumes were 332.1 ± 77.5 vs. 244.6 ± 63 vs. 417.9 ± 62.1 vs. 102.4 ± 18.5 for the saline (negative control), T-DM1 (positive control), T-PEG6-DM1, and [225Ac]Ac-macropa-T-PEG6-DM1, respectively. CONCLUSIONS [225Ac]Ac-macropa-T-PEG6-DM1 is more potent than ADC against trastuzumab-resistant breast cancer and necessitates clinical translation.
Collapse
Affiliation(s)
- Jessica Pougoue Ketchemen
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
- Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada
- Faculté de Pharmacie, Université Laval, Québec, Canada
| | - Fabrice Ngoh Njotu
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
- Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Hanan Babeker
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Alissar Monzer
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Emmanuel Nwangele
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
- Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Anjong Florence Tikum
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Nikita Henning
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Nava Hassani
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Sarah Frye
- Canadian Nuclear Laboratories, Radiobiology and Health Branch, Chalk River, Canada
| | - Randy Perron
- Canadian Nuclear Laboratories, Radiobiology and Health Branch, Chalk River, Canada
| | - Chris Byrne
- Canadian Nuclear Laboratories, Radiobiology and Health Branch, Chalk River, Canada
| | - Candice Didychuk
- Canadian Nuclear Laboratories, Radiobiology and Health Branch, Chalk River, Canada
| | - Qi Qi
- Canadian Nuclear Laboratories, Radiobiology and Health Branch, Chalk River, Canada
| | - Laura Bannister
- Canadian Nuclear Laboratories, Radiobiology and Health Branch, Chalk River, Canada
| | - Alireza Doroudi
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Humphrey Fonge
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
- Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Department of Medical Imaging, Royal University Hospital Saskatoon, Saskatoon, Canada
| |
Collapse
|
49
|
Chowdhury A, Shrestha P, Jois SD. Molecular Chimera in Cancer Drug Discovery: Beyond Antibody Therapy, Designing Grafted Stable Peptides Targeting Cancer. Int J Pept Res Ther 2025; 31:38. [PMID: 39974747 PMCID: PMC11832722 DOI: 10.1007/s10989-025-10690-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2025] [Indexed: 02/21/2025]
Abstract
Background Several cancer therapies are being developed, and given the variability of different cancer types, the goal of these therapies is to remove the invasive tumor from the body, kill the cancer cells, or else retard the growth. These include chemotherapeutic agents and targeted therapy using small molecules and antibodies. However, antibodies can generate an immune response upon repeated administration, and producing antibodies could be expensive. Purpose The purpose of this review is to describe different therapeutic approaches utilized for cancer therapy, the current therapeutic approaches, and their limitations. As a novel strategy to combat cancer, designing new stable peptide scaffolds such as cyclotides and sunflower trypsin inhibitors (SFTI) is described. Results Stable peptides that can target proteins can be used as therapeutic agents. Here, we review the utilization and amalgamation of plant-based peptides with biological epitopes in designing molecules called "Molecular Chimeras" using a grafted peptide strategy. These cyclic peptides can bind to target receptors or modulate protein-protein interactions as they bind with high affinity and selectivity. Grafted peptides also possess better serum stability owing to the head-to-tail cyclization and other structural modifications. Conclusion Stable cyclic peptides outweigh the other biologicals in terms of stability and manufacturing process. Peptides and peptidomimetics can be used as therapeutic agents, and these molecules provide alternatives for biologicals and small molecule inhibitors as drugs.
Collapse
Affiliation(s)
- Arpan Chowdhury
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University Baton Rouge, Skip Bertman Drive, Baton Rouge, LA-70803 USA
| | - Prajesh Shrestha
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University Baton Rouge, Skip Bertman Drive, Baton Rouge, LA-70803 USA
| | - Seetharama D. Jois
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University Baton Rouge, Skip Bertman Drive, Baton Rouge, LA-70803 USA
| |
Collapse
|
50
|
Zhang Y, Shang H, Zhang J, Jiang Y, Li J, Xiong H, Chao T. Drug Treatment Direction Based on the Molecular Mechanism of Breast Cancer Brain Metastasis. Pharmaceuticals (Basel) 2025; 18:262. [PMID: 40006075 PMCID: PMC11859690 DOI: 10.3390/ph18020262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Today, breast cancer (BC) is the most frequently diagnosed malignancy and a leading cause of cancer-related deaths among women worldwide. Brain metastases (BMs) are a common complication among individuals with advanced breast cancer, significantly impacting both survival rates and the overall condition of life of patients. This review systematically analyzes the innovative approaches to drug treatment for breast cancer brain metastases (BCBMs), with particular emphasis placed on treatments targeting molecular mechanisms and signaling pathways and drug delivery strategies targeting the blood brain barrier (BBB). The article discusses various drugs that have demonstrated effectiveness against BCBM, featuring a mix of monoclonal antibodies, nimble small-molecule tyrosine kinase inhibitors (TKIs), and innovative antibody-drug conjugates (ADCs). This study of various drugs and techniques designed to boost the permeability of the BBB sheds light on how these innovations can improve the treatment of brain metastases. This review highlights the need to develop new therapies for BCBM and to optimize existing treatment strategies. With a deeper comprehension of the intricate molecular mechanisms and advances in drug delivery technology, it is expected that more effective personalized treatment options will become available in the future for patients with BCBM.
Collapse
Affiliation(s)
- Yumin Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (H.S.); (Y.J.); (J.L.)
| | - Haotian Shang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (H.S.); (Y.J.); (J.L.)
| | - Jiaxuan Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Yizhi Jiang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (H.S.); (Y.J.); (J.L.)
| | - Jiahao Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (H.S.); (Y.J.); (J.L.)
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (H.S.); (Y.J.); (J.L.)
| | - Tengfei Chao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (H.S.); (Y.J.); (J.L.)
| |
Collapse
|