1
|
Zhang Y, Fan W, Su F, Zhang X, Du Y, Li W, Gao Y, Hu W, Zhao J. Discussion on the mechanism of HER2 resistance in esophagogastric junction and gastric cancer in the era of immunotherapy. Hum Vaccin Immunother 2025; 21:2459458. [PMID: 39875210 PMCID: PMC11776468 DOI: 10.1080/21645515.2025.2459458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/11/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is a critical biomarker and therapeutic target in gastric/gastroesophageal junction (G/GEJ) cancers, despite the initial success of HER2-targeted therapies, such as trastuzumab, resistance to these drugs has emerged as a major impediment to effective long-term treatment. This review examines the mechanisms of drug resistance in HER2-positive G/GEJ cancer, the primary mechanisms of resistance explored include alterations in the HER2 receptor itself, such as mutations and changes in expression levels, as well as downstream signaling pathways, and interactions with the tumor microenvironment (TME). Furthermore, the review discusses the Novel therapeutic approaches, including the use of antibody-drug conjugates (ADCs) and combination therapies are assessed for their potential to enhance outcomes. By integrating recent research findings and clinical trials, this review aims to provide oncologists and researchers with insights into developing more effective treatments for patients with drug-resistant HER2-positive G/GEJ cancer.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Graduate School, Changzhi Medical College, Changzhi, Shanxi, China
| | - Wenxuan Fan
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Graduate School, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Fei Su
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Graduate School, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoling Zhang
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Yunyi Du
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Weiling Li
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Graduate School, Changzhi Medical College, Changzhi, Shanxi, China
| | - Yangjun Gao
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Wenqing Hu
- Department of Gastrointestinal Surgery, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Jun Zhao
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| |
Collapse
|
2
|
Nam AR, Oh KS, Bang JH, Jeong Y, Choo SY, Kim HJ, Lee SI, Kim JM, Yoon J, Kim TY, Oh DY. YAP as a therapeutic target to reverse trastuzumab resistance. Gastric Cancer 2025:10.1007/s10120-025-01630-w. [PMID: 40542295 DOI: 10.1007/s10120-025-01630-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 05/29/2025] [Indexed: 06/22/2025]
Abstract
BACKGROUND Trastuzumab resistance in HER2-positive cancers remains a significant clinical challenge with limited therapeutic options. Although the tumor-promoting role of the Yes-associated protein (YAP) pathway is well established, its role in trastuzumab resistance remains unclear. METHODS We established four trastuzumab-resistant (HR) cell lines (NCI-N87HR, SNU216HR, SNU2670HR, and SNU2773HR) from HER2-positive gastric cancer and biliary tract cancer cell lines. YAP pathway activation was assessed using Phospho-RTK arrays, bulk RNA-Seq, and immunofluorescence. Antitumor effects of YAP targeting were evaluated with MTT assays, cell-cycle analysis, migration assays, RT-qPCR, ELISA, and xenograft models of SNU-2773 and SNU-2773HR cells. Immune modulation by YAP was studied through co-culture experiments with human PBMCs and cancer cells, followed by flow cytometry analysis of immune markers. RESULTS Upregulation and activation of the YAP/TAZ pathway were observed in HR cells, indicated by elevated ROR2 levels and nuclear translocation of YAP. This activation, driven by YAP/TEAD-dependent Wnt5a expression, suggests a positive-feedback mechanism that amplifies YAP activity. Elevated YAP and TEAD levels were observed in patient tumor tissues during disease progression following HER2-targeted therapies. Targeting YAP disrupted its oncogenic effects and restored sensitivity to trastuzumab, increased activation of CD4+ and CD8+ T cells in PBMCs, likely via PD-L1 downregulation and enhanced immunogenic cell death. Verteporfin, a YAP-TEAD inhibitor, effectively reduced tumor growth and increased apoptosis in mouse models bearing HR tumors. CONCLUSIONS Targeting the ROR2-YAP/TEAD axis presents a promising therapeutic approach to overcome trastuzumab resistance in HER2-positive cancers, offering a potential strategy for enhancing treatment efficacy and improving clinical outcomes.
Collapse
Affiliation(s)
- Ah-Rong Nam
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Kyoung-Seok Oh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Ju-Hee Bang
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Yoojin Jeong
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Sea Young Choo
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Hyo Jung Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Su In Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Jae-Min Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, 03080, Korea
| | - Jeesun Yoon
- Department of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Korea
| | - Tae-Yong Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Korea
| | - Do-Youn Oh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea.
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, 03080, Korea.
- Department of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Korea.
| |
Collapse
|
3
|
Goloudina A, Le Chevalier F, Authié P, Charneau P, Majlessi L. Shared neoantigens for cancer immunotherapy. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200978. [PMID: 40256120 PMCID: PMC12008704 DOI: 10.1016/j.omton.2025.200978] [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] [Indexed: 04/22/2025]
Abstract
Exploration of neoantigens holds the potential to be productive in immuno-oncotherapy. Among tumor-specific antigens, neoantigens result from genetic instability that gives rise to non-synonymous somatic mutations, highly specific to tumor cells. In addition to point mutations, gene rearrangements, indels leading to frameshifts, chromosomal translocations or inversions that may lead to fusion proteins, alternative mRNA splicing, and integration of genetic material of oncogenic viruses into the host genome provide consistent sources of neoantigens that are absent in healthy tissues. Out of these alterations, 2%-3% may generate T cell neoepitopes, possibly detectable by TCRs. Neoantigens are absent in healthy tissues and are thus at low risk of triggering autoimmunity. In addition, the host lymphocytes have not been rendered tolerant toward them and it is possible to induce immune responses against them. Here, we overview the two categories of neoantigens, i.e., private and shared, and their use in immuno-oncotherapy in selected pre-clinical and clinical studies. The vast majority of commonly occurring tumor-specific mutations are cancer causing and are permanently expressed by all malignant tumor cells, preventing the latter from escaping vaccine-induced anti-neoantigen immunity. The use of public neoantigens combined with efficient vaccine platforms can provide non-personalized "off-the-shelf" therapeutic vaccine candidates for broad-spectrum immunotherapy purposes.
Collapse
Affiliation(s)
- Anastasia Goloudina
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Fabien Le Chevalier
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Authié
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Charneau
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Laleh Majlessi
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| |
Collapse
|
4
|
Yalaza C, Antmen SE, Acuner SE, Oz H, Bulut E, Canacankatan N. Identification of novel HER2 ınhibitors: potential therapeutics for breast cancer. Discov Oncol 2025; 16:1122. [PMID: 40522572 DOI: 10.1007/s12672-025-02792-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 05/22/2025] [Indexed: 06/19/2025] Open
Abstract
Human epidermal growth factor receptor-2 (HER2) is a tyrosine kinase receptor involved in cell growth and differentiation. Targeting HER2 is a critical strategy in HER2-positive breast cancer treatment. Despite advancements in HER2-targeted therapies, drug resistance and side effects remain significant challenges. Therefore, identifying novel HER2 inhibitors with the potential to overcome resistance mechanisms while maintaining favorable drug-like properties is essential. Identifying novel HER2 inhibitors with high binding affinity and favorable drug-like properties is essential for overcoming these limitations. This study employed molecular docking and molecular dynamics simulations to evaluate the binding potential of plant-derived and synthetic compounds against HER2. The most promising candidates were further analyzed using ADMET profiling and binding free energy calculations to assess their drug-likeness and binding free energy. Among the tested compounds, axitinib, prunetin, and silymarin demonstrated strong HER2-binding affinities comparable to established inhibitors such as TAK-285 and lapatinib. Molecular dynamics simulations revealed that prunetin formed the most stable HER2-ligand complex, while axitinib exhibited the lowest binding free energy, indicating a strong interaction potential. ADMET analysis confirmed axitinib and prunetin as favorable drug candidates, whereas silymarin exhibited lower intestinal absorption. In conclusion, axitinib and prunetin emerged as promising HER2 inhibitors that may offer therapeutic advantages by addressing both drug resistance and toxicity concerns in HER2-positive breast cancer treatment. Prunetin, with its lower toxicity and higher stability, presents a safer therapeutic option, whereas axitinib offers high binding affinity. These findings suggest that these compounds could help overcome resistance and side effects associated with current HER2-targeted therapies.
Collapse
Affiliation(s)
- Cem Yalaza
- Department of Biochemistry, Faculty of Pharmacy, Başkent University, Ankara, Turkey.
| | - Serife Efsun Antmen
- Department of Biochemistry, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Saliha Ece Acuner
- Science and Advanced Technologies Research Center (BILTAM), Department of Bioengineering, Istanbul Medeniyet University, Istanbul, Turkey
| | - Hasan Oz
- Mersin Public Health Laboratory, Mersin, Turkey
| | - Ecem Bulut
- Science and Advanced Technologies Research Center (BILTAM), Department of Bioengineering, Istanbul Medeniyet University, Istanbul, Turkey
| | - Necmiye Canacankatan
- Department of Biochemistry, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| |
Collapse
|
5
|
Long Y, Tang B, Xie F, Liu L, Zhou Y, Dong J, Wang J, Sun C, Wang Y, Li R, Zhang N, Li L, Luo L, Xiao J, Zhong W, Xiao D, Deng H, Zhou X. Novel Quaternary Ammonium Salt-Linked STING Agonist Antibody-Drug Conjugate: Synergistic Activation of Tumor Immunity with Mitigated Off-Target Toxicity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e02270. [PMID: 40492586 DOI: 10.1002/advs.202502270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 05/10/2025] [Indexed: 06/12/2025]
Abstract
Immune-stimulating antibody conjugate (ISACs) incorporating STING agonists as payloads leverage both the targeting capability of the Fab region and the Fc region-mediated tumor antigen-dependent immune activation. Herein, a novel class of ISACs is reported, generated by engineering a quaternary ammonium-cleavable linker to conjugate diABZI STING agonist 3 (dSA3) with the HER2-targeting antibody Trastuzumab. The optimized ISAC (TZ-dSA3-12) demonstrated high potency, stability, enhanced solubility, and reduced off-target toxicity. The data showed that TZ-dSA3-12 potently activates the STING pathway in the tumor microenvironment through the synergistic action of the Fab and Fc regions of antibodies (activity switch-on). In contrast, TZ-dSA3-12 exhibited ≈75 fold lower activity than dSA3 in normal immune cells, where activation relies solely on the Fc region without Fab-mediated tumor antigen binding (activity switch-off). Furthermore, systemic administration of TZ-dSA3-12 at a dose (1 mg kg-1) elicited robust and sustained antitumor effect in a manner dependent on the activation of innate immunity and adaptive immunity, including macrophages, dendritic cells (DCs) and CD8+ T cells, while minimizing systemic cytokine release. Notably, TZ-dSA3-12 also induced immunological memory to combat the growth of rechallenged tumors. This innovative quaternary ammonium-linked STING agonist-ISAC represents a promising avenue for the future development of STING-targeted immunotherapy.
Collapse
Affiliation(s)
- Yu Long
- College of Pharmacy, Qingdao University, Qingdao, 266071, China
| | - Borui Tang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Fei Xie
- National Engineering Research Center for the Emergency Drug, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Lianqi Liu
- National Engineering Research Center for the Emergency Drug, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Yangyihua Zhou
- Academy of Military Medical Sciences, Beijing, 100850, China
| | - Jingwen Dong
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jianfeng Wang
- National Engineering Research Center for the Emergency Drug, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Cuicui Sun
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yuting Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Ruoqi Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Na Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Liping Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Longlong Luo
- Academy of Military Medical Sciences, Beijing, 100850, China
| | - Junhai Xiao
- National Engineering Research Center for the Emergency Drug, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Dian Xiao
- National Engineering Research Center for the Emergency Drug, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Hongbin Deng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Xinbo Zhou
- National Engineering Research Center for the Emergency Drug, Academy of Military Medical Sciences, Beijing, 100850, China
| |
Collapse
|
6
|
Ma D, Dai LJ, Wu XR, Liu CL, Zhao S, Zhang H, Chen L, Xiao Y, Li M, Zhao YZ, Yang L, Zhou T, Li JJ, Yang WT, Jiang YZ, Shao ZM. Spatial determinants of antibody-drug conjugate SHR-A1811 efficacy in neoadjuvant treatment for HER2-positive breast cancer. Cancer Cell 2025; 43:1061-1075.e7. [PMID: 40215979 DOI: 10.1016/j.ccell.2025.03.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/17/2024] [Accepted: 03/12/2025] [Indexed: 06/18/2025]
Abstract
Selecting optimal candidates for next-generation anti-human epidermal growth factor receptor 2 (HER2) antibody-drug conjugates (ADCs) remains challenging. We conduct a prespecified translational study to identify treatment biomarkers in SHR-A1811-treated HER2-positive breast cancer patients from the phase 2 neoadjuvant FASCINATE-N trial using DNA and RNA sequencing, computational pathology, and single-cell in situ spatial imaging. In the hormone receptor (HR)-negative subgroup, a higher proportion and more infiltration of immune cells (i.e., tumor-infiltrating lymphocytes [TILs]), particularly cytotoxic T cells, are associated with better treatment responses. In the HR-positive subgroup, the closeness and aggregation of HER2-strong-positive tumor cells, as opposed to a uniform distribution, are linked to a lower response rate and HER2 luminal-like (HER2-LUM) subtype, which more closely resembles HR+/HER2- breast cancer. In addition, we develop a clinically practical predictive model capable of predicting neoadjuvant treatment responses to SHR-A1811 and other novel ADCs based on clinicopathological characteristics and pathological images.
Collapse
Affiliation(s)
- Ding Ma
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lei-Jie Dai
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiang-Rong Wu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Cheng-Lin Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shen Zhao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hang Zhang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Li Chen
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi Xiao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ming Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yi-Zhi Zhao
- School of Engineering, Westlake University, Hangzhou 310030, China; Hangzhou DiPath Technology Co. Ltd., Hangzhou 311121, China
| | - Lin Yang
- School of Engineering, Westlake University, Hangzhou 310030, China; Hangzhou DiPath Technology Co. Ltd., Hangzhou 311121, China
| | - Tong Zhou
- Shanghai Institute of Preventive Medicine, Shanghai 201416, China
| | - Jun-Jie Li
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Wen-Tao Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Academy of Natural Sciences (SANS), Fudan University, Shanghai 200030, China.
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
7
|
Lee SH, Lee H, Kwon YJ, Kim SK, Seo EB, Sohn JO, Kim BH, Park JY, Ye SK. Chalcone-9: a novel inhibitor of the JAK-STAT pathway with potent anti-cancer effects in triple-negative breast cancer cells. Pharmacol Rep 2025; 77:761-774. [PMID: 40199813 PMCID: PMC12066378 DOI: 10.1007/s43440-025-00721-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Breast cancer remains the leading cause of cancer incidence and mortality among women worldwide, with triple-negative breast cancer (TNBC) posing significant treatment challenges. The dysregulation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway contributes to tumor progression, making it a potential therapeutic target. Chalcones, known for their diverse biological activities, including anti-cancer effects, hold promise for drug development. This study explores the anti-cancer activity of (E)-4-(3-(2-(benzyloxy)-6-hydroxyphenyl)-3-oxoprop-1-en-1-yl)benzoic acid (chalcone-9), a novel chalcone derivative. METHODS The cytotoxic effects of chalcone-9 were evaluated in breast cancer cell lines, including TNBC lines MDA-MB-231 and MDA-MB-468. Western blotting and qRT-PCR were used to analyze the impact on JAK1, JAK2, STAT1, and STAT3 activation and their downstream gene expression. In silico molecular docking analysis was conducted to determine whether chalcone-9 can interact with JAK1 and JAK2. A wound healing assay was used to observe the effect of chalcone-9 on tumor cell migration, and flow cytometry was employed to analyze whether chalcone-9 inhibits tumor cell cycle progression and induces apoptosis. The expression of apoptosis markers was also assessed. RESULTS Chalcone-9 exhibited dose-dependent cytotoxicity in breast cancer cell lines, with TNBC cells showing higher sensitivity. Chalcone-9 effectively inhibited the activation of JAK1, JAK2, STAT1, and STAT3, outperforming conventional JAK/STAT inhibitors. The structure of chalcone-9 was confirmed to stably interact with JAK1 and JAK2 proteins. It also suppressed STAT1 and STAT3 target gene expression, reduced tumor cell migration, and induced apoptosis, as evidenced by PARP and caspase cleavage and decreased survivin levels. CONCLUSIONS Chalcone-9 demonstrates significant anti-cancer activity, particularly against TNBC. By targeting the JAK/STAT pathway and promoting apoptosis, chalcone-9 emerges as a promising therapeutic candidate for aggressive breast cancers.
Collapse
Affiliation(s)
- Song-Hee Lee
- Department of Biomedical Sciences and Pharmacology, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
| | - Haeri Lee
- Department of Biomedical Sciences and Pharmacology, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Yong-Jin Kwon
- Department of Cosmetic Science, Kyungsung University, Busan, 48434, Republic of Korea
| | - Seul-Ki Kim
- Department of Biomedical Sciences and Pharmacology, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
| | - Eun-Bi Seo
- Department of Biomedical Sciences and Pharmacology, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jie Ohn Sohn
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Gangwon-Do, 25159, Republic of Korea
| | - Byung-Hak Kim
- Medience Co. Ltd., Chuncheon, Gangwon-Do, 24232, Republic of Korea
| | - Jung-Youl Park
- Glocal University Project Group, Andong National University, Andong, Gyeongsangbuk-Do, 36729, Republic of Korea
| | - Sang-Kyu Ye
- Department of Biomedical Sciences and Pharmacology, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Gangwon-Do, 25159, Republic of Korea.
- Biomedical Science Project (BK21PLUS), Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Neuro-Immune Information Storage Network Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
8
|
Liu D, Liu L, Zhang X, Zhao X, Li X, Che X, Wu G. Decoding driver and phenotypic genes in cancer: Unveiling the essence behind the phenomenon. Mol Aspects Med 2025; 103:101358. [PMID: 40037122 DOI: 10.1016/j.mam.2025.101358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/25/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Gray hair, widely regarded as a hallmark of aging. While gray hair is associated with aging, reversing this trait through gene targeting does not alter the fundamental biological processes of aging. Similarly, certain oncogenes (such as CXCR4, MMP-related genes, etc.) can serve as markers of tumor behavior, such as malignancy or prognosis, but targeting these genes alone may not lead to tumor regression. We pioneered the name of this class of genes as "phenotypic genes". Historically, cancer genetics research has focused on tumor driver genes, while genes influencing cancer phenotypes have been relatively overlooked. This review explores the critical distinction between driver genes and phenotypic genes in cancer, using the MAPK and PI3K/AKT/mTOR pathways as key examples. We also discuss current research techniques for identifying driver and phenotypic genes, such as whole-genome sequencing (WGS), RNA sequencing (RNA-seq), RNA interference (RNAi), CRISPR-Cas9, and other genomic screening methods, alongside the concept of synthetic lethality in driver genes. The development of these technologies will help develop personalized treatment strategies and precision medicine based on the characteristics of relevant genes. By addressing the gap in discussions on phenotypic genes, this review significantly contributes to clarifying the roles of driver and phenotypic genes, aiming at advancing the field of targeted cancer therapy.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Lei Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xiaoman Zhang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xinming Zhao
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xiaorui Li
- Department of Oncology, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, China.
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
9
|
Shen K, Yuan S, Su N, Tang F, Rehim S, Wang H, Guo H, Zhang Y, Wu Y, Wang H. Monotherapy and combination therapy using antibody‑drug conjugates for platinum‑resistant ovarian cancer. Oncol Rep 2025; 53:68. [PMID: 40242965 PMCID: PMC12046379 DOI: 10.3892/or.2025.8901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Platinum‑resistant ovarian cancer (PROC) is a significant clinical challenge due to the limited number of treatment options and poor outcomes. Moreover, cytotoxic drugs have an unsatisfactory therapeutic efficacy, high toxicity and side effects. An antibody‑drug conjugate (ADC) is a novel cancer therapeutic strategy that combines an antibody, a linker and a payload. ADCs precisely target the tumor cells by binding to the antigen on the surface of tumor cells, thus accurately delivering the cytotoxic drugs and minimizing systemic toxicity. The approval of mirvetuximab soravtansine by the US Food and Drug Administration for treating folate receptor alpha‑positive, platinum‑resistant epithelial ovarian cancer has promoted studies on the use of ADCs in ovarian cancer. A phase III clinical trial showed that mirvetuximab soravtansine achieved an objective remission rate of 42.3% in platinum‑resistant, FRα‑positive ovarian cancer, compared with 15.9% using chemotherapy, demonstrating its immense potential for ADC development. The present review summarizes the research progress on the use of ADCs in PROC as a monotherapy and combination therapy and considers the future development direction of ADCs in PROC.
Collapse
Affiliation(s)
- Ke Shen
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Shuang Yuan
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Ning Su
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
- Department of Gynecologic Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Furong Tang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Shamsnur Rehim
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Han Wang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Huihui Guo
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Yu Zhang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Yufeng Wu
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Hongjing Wang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
10
|
Zhang M, Yang Q, Lou J, Hu Y, Shi Y. A new strategy to HER2-specific antibody discovery through artificial intelligence-powered phage display screening based on the Trastuzumab framework. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167772. [PMID: 40056877 DOI: 10.1016/j.bbadis.2025.167772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a recognized drug target, and it serves as a critical target for various cancer treatments, necessitating the discovery of more antibodies for therapeutic and detection purposes. Here, we have developed an innovative workflow for antibody generation through Artificial Intelligence-powered Phage Display Screening (AIPDS). This workflow integrates artificial intelligence-driven antibody CDRH3 sequence design, high-throughput DNA synthesis and phage display screening. We applied AIPDS workflow to generate promising antibodies against the human epidermal growth factor receptor 2 (HER2), offering a template for streamlined antibody generation. Seven novel antibodies stood out, demonstrating promising efficacy in various functional assays. Notably, DYHER2-02 demonstrates strong performance across all experimental tests. In summary, our study introduces a novel methodology to generate new antibody variants of an existing antibody using an AI-assisted phage display approach. These new antibody variants hold potential applications in research, diagnosis, and therapeutic applications.
Collapse
Affiliation(s)
- Mancang Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Qiangzhen Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Jiangrong Lou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Yang Hu
- United Research Center for Next Generation DNA Synthesis of SJTU-Dynegene, Shanghai 201108, People's Republic of China
| | - Yongyong Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China; Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China.
| |
Collapse
|
11
|
Liu J, Liu J, Yu J, Ren Q, Cai Y, Chen D, Song C. Research advancements of antibody drug conjugates in non-small cell lung cancer with HER2 alterations. J Transl Med 2025; 23:600. [PMID: 40448190 DOI: 10.1186/s12967-025-06589-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 05/07/2025] [Indexed: 06/02/2025] Open
Abstract
The human epidermal growth factor receptor 2 (HER2) alterations are significant genetic alterations in non-small cell lung cancer (NSCLC), encompassing mutations, amplifications, and protein overexpression. Despite the substantial progress of anti-HER2 targeted therapies in breast and gastric cancers, numerous challenges persist in the treatment of NSCLC with HER2 alterations. Presently, the options for NSCLC with HER2 alterations remain limited, with inferior efficacy observed using small molecule anti-tumor targeted agents and conventional chemotherapy. Antibody drug conjugates (ADCs), an organic combination of monoclonal antibodies and cytotoxic drugs targeting specific tumor cells, have revolutionized the treatment landscape of NSCLC with HER2 alterations. Extensive exploration of ADCs has been conducted across NSCLC patients with HER2 alterations, achieving notable efficacy in some populations. This review aims to delve into the biological characteristics and current treatment landscape of NSCLC with HER2 alterations, emphasizing the transformative research advancements surrounding ADCs. By highlighting these developments, we aspire to provide essential insights to enhance clinical practice and improve management strategies for NSCLC patients with HER2 alterations.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Oncology, Xinghua People's Hospital Affiliated to Yangzhou University, 419 Ying Wu Nan Road, Xinghua, 225700, Jiangsu, People's Republic of China.
| | - Jianhua Liu
- Department of Oncology, Xinghua People's Hospital Affiliated to Yangzhou University, 419 Ying Wu Nan Road, Xinghua, 225700, Jiangsu, People's Republic of China
| | - Jianhe Yu
- Department of Oncology, Xinghua People's Hospital Affiliated to Yangzhou University, 419 Ying Wu Nan Road, Xinghua, 225700, Jiangsu, People's Republic of China
| | - Qun Ren
- Department of Oncology, Xinghua People's Hospital Affiliated to Yangzhou University, 419 Ying Wu Nan Road, Xinghua, 225700, Jiangsu, People's Republic of China
| | - Yin Cai
- Department of Oncology, Xinghua People's Hospital Affiliated to Yangzhou University, 419 Ying Wu Nan Road, Xinghua, 225700, Jiangsu, People's Republic of China
| | - Dadong Chen
- Department of Oncology, Xinghua People's Hospital Affiliated to Yangzhou University, 419 Ying Wu Nan Road, Xinghua, 225700, Jiangsu, People's Republic of China
| | - Chuanjun Song
- Department of Oncology, Xinghua People's Hospital Affiliated to Yangzhou University, 419 Ying Wu Nan Road, Xinghua, 225700, Jiangsu, People's Republic of China.
| |
Collapse
|
12
|
Zhou H, Shi M, Yang W, Song N, Luo X. Engineered Antifouling Peptides with Sarcosine Branches for Robust Electrochemical Detection of the HER2 Biomarker in Real Biological Samples. ACS Sens 2025. [PMID: 40415312 DOI: 10.1021/acssensors.5c00082] [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: 05/27/2025]
Abstract
In complex biological matrices, the nonspecific adsorption phenomena occurring on the surfaces of electrochemical biosensors represent a considerable challenge for the precise detection of targets in heterogeneous biological samples. Furthermore, the presence of protein hydrolases in biofluids also affects the stability of biosensing devices utilizing natural proteins or peptides. It is therefore imperative to develop sensing devices capable of effectively minimizing such effects in real biological samples. Herein, we engineered a sarcosine branch-chain peptide (SBCP) with a strong antifouling capability to avoid biofouling and enhanced stability to resist hydrolysis by proteases. The peptide is composed of three sections: an anchoring sequence (CPPPP), an antifouling sequence (EK(Sar)EK(Sar)EK(Sar)EK(Sar)), and a recognition sequence (HLTVSPWY). An electrochemical biosensor was developed through the electrodeposition of poly(3,4-ethylenedioxythiophene) (PEDOT) incorporated with poly(norepinephrine) (PNE) on an electrode surface, followed by the electrodeposition of gold nanoparticles and the self-assembly of SBCP. The biosensor constructed using the SBCP containing a specific recognizing peptide sequence for the cancer biomarker human epidermal growth factor receptor 2 (HER2) was capable of sensitively detecting target HER2, within the concentration range of 1.0 pg mL-1 to 1.0 μg mL-1 and with a limit of detection of 0.37 pg mL-1. Moreover, the biosensor demonstrated antifouling ability and the capacity to accurately detect the target in human serum, exhibiting a high degree of concordance with the assaying results of ELISA kits. These findings suggest that the biosensor based on the engineered peptides possesses promising potential for practical applications.
Collapse
Affiliation(s)
- Hao Zhou
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; Shandong Key Laboratory of Biochemical Analysis; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P.R. China
| | - Mingjun Shi
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; Shandong Key Laboratory of Biochemical Analysis; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P.R. China
| | - Wenhao Yang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; Shandong Key Laboratory of Biochemical Analysis; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P.R. China
| | - Ning Song
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; Shandong Key Laboratory of Biochemical Analysis; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P.R. China
| | - Xiliang Luo
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; Shandong Key Laboratory of Biochemical Analysis; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P.R. China
| |
Collapse
|
13
|
Guo X, Xu C, Zhu X, Wang X, Hao Y, Wang X, Yao Y, Fang J, Wang K. In Silico Design and Evaluation of Novel Peptide-Based PET Probe for Noninvasive Imaging of HER2 Expression in Breast Cancer. J Med Chem 2025; 68:10461-10472. [PMID: 40322890 DOI: 10.1021/acs.jmedchem.5c00760] [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: 05/23/2025]
Abstract
The overexpression of HER2 has been strongly correlated with the pathogenesis and progression of breast cancer. Developing HER2-targeted PET tracers for noninvasive assessment of HER2 expression holds substantial clinical significance for screening patients suitable for HER2-targeted therapy, evaluating treatment efforts, and optimizing treatment. In this study, we employed computational simulation methods to develop peptide WC8 derived from trastuzumab, and designed HER2 targeting radiotracers [68Ga]Ga-DOTA-WC8 for detecting the HER2 expression. The radiotracer demonstrated nanomolar affinity, high specificity, and favorable pharmacokinetics. PET imaging revealed that [68Ga]Ga-DOTA-WC8 exhibited significant and specific uptake in HER2-positive breast cancer tissues, facilitating the rapid and accurate identification of HER2 expression. Notably, this radiotracer was successfully utilized to achieve direct visualization of tumor response to pyrotinib, a HER2 tyrosine kinase inhibitor. These findings suggest that the peptide-based radiotracer [68Ga]Ga-DOTA-WC8 represents a promising tool for accurately monitoring of HER2 expression after treatment with HER2-targeting drugs.
Collapse
Affiliation(s)
- Xu Guo
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
- Department of Pharmacy, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu 214002, China
| | - Chunwei Xu
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xue Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiaojia Wang
- Department of Breast Medical Oncoloy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Yue Hao
- Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China
| | - Xun Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Ying Yao
- Department of Pharmacy, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu 214002, China
| | - Jing Fang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ke Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
14
|
Wang A, Wang X, Li D, Li A, He M, Yuan Y, Ye L, Liu J. A superior method for antitumor therapy and application: dual-ligand nanomedicines. J Mater Chem B 2025. [PMID: 40396464 DOI: 10.1039/d5tb00044k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Currently, nanomedicines have been widely applied in the treatment of various types of tumors. However, due to the complexity of the tumor microenvironment, conventional nanomedicines often exhibit poor efficacy, insufficient site specificity, and susceptibility to off-target effects. In contrast, dual-ligand nanomedicines demonstrate superior targeting ability and drug penetration in tumor therapy. These nanomedicines are equipped with two ligands on their surface, enabling targeting of specific receptors on the same or different cells. The specific binding between ligands and receptors significantly enhances the selectivity and targeting of dual-ligand nanomedicines towards tumors. This review systematically describes the preparation of dual-ligand nanomedicines, the influencing factors, and the types of delivered drugs, focusing on the application of dual-ligand nanomedicines in targeting the treatment of various tumors. We highlight the comprehensiveness of dual-ligand nanomedicines for the treatment of tumors, including glioblastoma, lung cancer, breast cancer, gastric cancer, and many other types of tumors. Finally, the possible challenges for the future development of dual-ligand nanomedicines in terms of preparation, clinic, and safety are further analyzed. We look forward to exploring dual-ligand nanomedicines in greater depth to provide references for their future development and clinical applications.
Collapse
Affiliation(s)
- Ailing Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Xuejun Wang
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Dan Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Aixue Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Mengyuan He
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| | - Yingying Yuan
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
- College of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Ye
- School of Pharmacy and (R & D Center) Lab. for Drug Discovery from Natural Resource, Macau University of Science and Technology, Macau SAR, 999078, China.
| | - Jiyong Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210000, China
| |
Collapse
|
15
|
Hashemi SMJ, Ghalehnoei H, Barzegar A, Feizi-Dehnayebi M, Akhtari J, Mellati A. In silico discovery of multi-target small molecules and efficient siRNA design to overcome drug resistance in breast cancer via local therapy. J Mol Graph Model 2025; 140:109086. [PMID: 40424842 DOI: 10.1016/j.jmgm.2025.109086] [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: 05/14/2025] [Accepted: 05/18/2025] [Indexed: 05/29/2025]
Abstract
In this study, we designed an efficient siRNA for PKMYT1 gene knockdown and evaluated the binding affinity of various natural small molecules to key proteins associated with breast cancer through molecular docking and molecular dynamics (MD) simulations. Subsequently, among these molecules, The small molecule, SCHEMBL7562664, was introduced as a "golden ligand" that showed potent multi-target activity as an antagonist for aromatase, estrogen receptor α, HER2, and PARP10, and as an agonist for MT2 and STING. Next, MD simulations of six protein- golden ligand complexes (PDB IDs: 4QXQ, 5GS4, 5JL6, 5LX6, 6ME6, and 7PCD), performed with GROMACS over 100 ns at 298.15 K, provided valuable information about their structural dynamics. Analysis of the radius of gyration (Rg) revealed that, while five complexes (7PCD, 5GS4, 5LX6, 4QXQ, and 5JL6) maintained compact structures (Rg between 1.7 and 2.3 nm), the 6ME6 complex exhibited a more extended and flexible conformation (average Rg ∼3.4 nm). Complementary RMSD analysis confirmed that most complexes rapidly stabilized with minimal deviations (generally <0.3 nm), whereas the 6ME6 complex showed higher variability, reaching up to 0.67 nm. Furthermore, Binding free energy calculations using MM-GBSA and PBSA methods further supported these findings, with energies ranging from -21.45 ± 2.28 kcal/mol (5LX6) to -39.79 ± 1.34 kcal/mol (6ME6), indicating an optimal balance between intrinsic interactions and desolvation costs in the 6ME6 and 5JL6 systems. Based on DFT results, the golden ligand showed higher stability and lower reactivity compared to control ligands such as aromatase, tamoxifen, and dacomitinib, potentially leading to reduced off-target interactions and a more favorable safety profile. The integration of these data underscores the therapeutic potential of SCHEMBL7562664 as a multi-target agent for breast cancer, with promising pharmacokinetic properties that can be optimized for local treatment by incorporation into a 3D scaffold.
Collapse
Affiliation(s)
| | - Hossein Ghalehnoei
- Department of Medical Biotechnology, Molecular and Cell Biology Research Center, Faculty of Advanced Technologist in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Barzegar
- Department of Basic Sciences, Sari Agricultural Sciences and Natural Resources University, Sari, Iran
| | - Mehran Feizi-Dehnayebi
- Department of Organic Chemistry, Faculty of Chemistry, Alzahra University, Tehran, Iran.
| | - Javad Akhtari
- Immunogenetics Research Center, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amir Mellati
- Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
16
|
Zidel A, Benton A, Brown E, Shmuel S, Vanover A, Panikar SS, Bose R, Park H, Davis AA, Pereira PMR. Modulation of HER2 internalization enhances single-dose antibody-drug potency in HER2 + gastric cancer. Sci Rep 2025; 15:16964. [PMID: 40374836 PMCID: PMC12081668 DOI: 10.1038/s41598-025-01947-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 05/09/2025] [Indexed: 05/18/2025] Open
Abstract
HER2 is a membrane receptor tyrosine kinase overexpressed in 18-20% of gastric tumors. Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate (ADC) that targets HER2-positive (HER2+) cancer cells with a chemotherapeutic agent, emtansine. T-DM1 has low efficacy in HER2+ gastric cancer. This study explored the efficacy of combining drugs known to modulate HER2 internalization to enhance T-DM1 efficacy in gastric cancer. We used cholesterol-depleting drugs (lovastatin) to enhance HER2 membrane density. The irreversible pan-HER tyrosine kinase inhibitor neratinib was used to enhance the internalization of HER2-bound T-DM1. Therapy, pre-treatment and post-treatment positron emission tomography/computed tomography (PET/CT) were performed in both male and female mice. An enhancement of cell surface and internalized HER2 was observed after lovastatin and neratinib incubations, respectively. The combination of lovastatin with neratinib enhanced T-DM1 internalization in cancer cells. A decrease in HER2 protein levels and HER2 phosphorylation was detected in cells treated with T-DM1/lovastatin/neratinib when compared to control and T-DM1-only groups. PET/CT imaging of mice in the T-DM1/lovastatin/neratinib group showed a decrease in HER2 tumoral expression, which was associated with a decrease in tumor volume and sustained treatment efficacy in the T-DM1/lovastatin/neratinib group. This work demonstrates the therapeutic enhancement of T-DM1 using combination therapy with lovastatin/neratinib in gastric cancer. The treatment can be successfully monitored through PET/CT imaging.
Collapse
Affiliation(s)
- Abbey Zidel
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Alex Benton
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Cancer Biology Graduate Program, Washington University School of Medicine, St. Louis, MO, USA
| | - Emma Brown
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Shayla Shmuel
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Alex Vanover
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sandeep Surendra Panikar
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ron Bose
- Washington University School of Medicine, 4515 McKinley Avenue, Campus Box 8069, St. Louis, MO, USA
| | - Haeseong Park
- Gastrointestinal Cancer Center, Center for Cancer Therapeutic Innovation, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew A Davis
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Patrícia M R Pereira
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
17
|
Xu H, Liang Y, Tang W, Yang X, Du X. Clinical significance of HER2 overexpression in biliary tract carcinoma --a meta analysis. Front Oncol 2025; 15:1534005. [PMID: 40438678 PMCID: PMC12116302 DOI: 10.3389/fonc.2025.1534005] [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: 11/25/2024] [Accepted: 04/16/2025] [Indexed: 06/01/2025] Open
Abstract
Introduction Previous studies have been inconsistent on the correlation of human epidermal growth factor receptor 2 (HER2) overexpression in biliary tract carcinoma. The objective of this meta analysis was to assess its association with clinicopathological features and prognostic significance of biliary tract carcinoma. Methods The eligible studies were searched in Pubmed, Embase and Web of Science databases. Inclusion criteria were studies of the relationship between HER2 positive expression (ICH: HER2 (+++), FISH: HER2 overexpression, NGS: HER2 overexpression) and prognosis or clinicopathological features of patients with biliary tract carcinoma (BTC). The analysis was conducted according to gender, high differentiation degree, middle differentiation degree, tumor stage, nerve invasion, vascular invasion, lymph node metastasis and pathological diagnosis of patients. ES (Effect Sizes) for 95% confidence intervals (CI) were calculated to examine risk or hazard associations, and heterogeneity and sensitivity analyses were performed. Results A total of 15 studies were included to evaluate the association of HER2 positive expression with clinicopathological features and survival prognosis. There was no significant statistical relationship between positive/high expression of HER2 and a series of clinical characteristics including gender, high, middle and low differentiation, tumor stage, vascular invasion, nerve invasion, Lymph node metastasis, T stage and pathological type of patients with biliary tract carcinoma. There was a significant relationship between positive/high expression of HER2 and postoperative Disease-Free Survival in patients with biliary tract carcinoma (ES = 1.87, 95% CI: 1.24-2.81, p = 0.003). There was a significant relationship between positive/high expression of HER2 and postoperative Overall Survival in patients with biliary tract carcinoma (ES = 1.54, 95% CI: 1.08-2.20, p = 0.017). Discussion Our meta-analyses revealed that high expression of HER2 gene was not correlated with clinicopathological parameters such as differentiation degree, TNM stage, lymph node metastasis, vascular invasion, nerve invasion, pathological type, T stage, and gender of biliary tract carcinoma. HER2 overexpression is a negative prognostic factor in biliary tract carcinoma patients. The association between positive/high expression of HER2 and the pathological features as well as prognosis in biliary tract carcinoma patients warrants further validation.
Collapse
Affiliation(s)
- Haonan Xu
- Department of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, Sichuan, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, Mianyang, Sichuan, China
| | - Yuwen Liang
- Department of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, Sichuan, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, Mianyang, Sichuan, China
| | - Wenqiang Tang
- Department of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, Sichuan, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, Mianyang, Sichuan, China
| | - Xiongxin Yang
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xiaobo Du
- Department of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, Sichuan, China
- Sichuan Clinical Research Center for Radiation and Therapy, Mianyang Central Hospital, Mianyang, Sichuan, China
| |
Collapse
|
18
|
Zhang X, Zhao J, Yin X, Liang J, Wang Y, Zheng L, Tan P, Lin Y, Xu N, Zhu S, Chen J, Zhao J, Hu X, Pan X, Nie L, Zhang M, Chen Y, Zhang Y, Liu H, Dai J, Wang Z, Liu H, Ni Y, Rupp NJ, Moch H, Sheng X, Gong K, Liu X, Chen Z, He Z, Wang Y, Xu L, Liu M, Zhou H, Tang B, Huang R, Wei Q, Li X, Liu J, Yao J, Liao B, Liu Z, Shen P, Chen N, Zeng H, Sun G. Comprehensive molecular profiling of FH-deficient renal cell carcinoma identifies molecular subtypes and potential therapeutic targets. Nat Commun 2025; 16:4398. [PMID: 40355427 PMCID: PMC12069531 DOI: 10.1038/s41467-025-59513-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/25/2025] [Indexed: 05/14/2025] Open
Abstract
Fumarate hydratase-deficient renal cell carcinoma (FH-deficient RCC) is a rare yet highly lethal kidney cancer. To deepen our understanding of FH-deficient RCC, we conduct a comprehensive integrated genomic study. We analyze the association of FH alteration patterns with tumor heterogeneity and develop a CpG site-specific methylation signature for precise identification of FH-deficient RCC. Transcriptomic analysis unveils three distinctive molecular subtypes characterized by enrichment of immune/Angiogenic/Stromal (C1), WNT/Notch/MAPK (C2), and proliferation/stemness (C3) pathways, respectively. Tumors in C1 derive the most substantial survival benefit from a combination of immune checkpoint blockade (ICB) and anti-angiogenic therapy. Tumors in C2 display moderate response to this therapeutic approach. In contrast, tumors in C3 exhibit an unfavorable response to anti-angiogenic monotherapy and its combination with ICB. These findings contribute to a profound understanding of the aggressive nature of FH-deficient RCC, offering insights into potential precision medicine approaches for disease management.
Collapse
Affiliation(s)
- Xingming Zhang
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Junjie Zhao
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxue Yin
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiayu Liang
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Yongquan Wang
- Department of Urology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Linmao Zheng
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Ping Tan
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Yifei Lin
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
- Department of Urology, Medical Device Regulatory Research and Evaluation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Nanwei Xu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Sha Zhu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Junru Chen
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Jinge Zhao
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Xu Hu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Xiuyi Pan
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Nie
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Mengni Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuntian Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yaowen Zhang
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Haoyang Liu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Jindong Dai
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Zhipeng Wang
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Haolin Liu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Yuchao Ni
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, Zurich, CH-8006, University of Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, Zurich, CH-8006, University of Zurich, Zurich, Switzerland
| | - Xinan Sheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genitourinary Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Kan Gong
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Xiaodong Liu
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhibin Chen
- Department of Urology, The First People's Hospital of Neijiang, Neijiang, China
| | - Zhengyu He
- Department of Urology, Yaan People's Hospital, Yaan, China
| | - Yaodong Wang
- Department of Urology, Mianyang Central Hospital, Mianyang, China
| | - Lijing Xu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Mingsheng Liu
- The Second Ward of Urology, Affiliated Qujing Hospital of Kunming Medical University, Qujing, China
| | - Hongqing Zhou
- The Second Ward of Urology, Affiliated Qujing Hospital of Kunming Medical University, Qujing, China
| | - Bo Tang
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Huang
- Department of Nuclear medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Li
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Jiyan Liu
- Department of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Yao
- Department of Urology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Banghua Liao
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Pengfei Shen
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China.
| | - Ni Chen
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China.
| | - Hao Zeng
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China.
| | - Guangxi Sun
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
19
|
Zhou X, Xiao L, Lai F, Chen W, Zhou C, Deng Y, Wang T, Xing S, Diao H, Tang M, Guo W, Luo E. Comprehensive overview of antibody drug-related clinical studies in gynecology: insights from ClinicalTrials.gov. Front Med (Lausanne) 2025; 12:1521587. [PMID: 40417700 PMCID: PMC12098050 DOI: 10.3389/fmed.2025.1521587] [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: 11/02/2024] [Accepted: 04/18/2025] [Indexed: 05/27/2025] Open
Abstract
Antibodies have been widely used globally over the past decade and play an increasingly important role in modern medicine. Notably, significant advancements have been achieved in the realm of gynecology, particularly in gynecological cancers. This study endeavors to present a thorough overview of antibody-related drug clinical studies in gynecology registered on ClinicalTrials.gov, focusing on the basic characteristics of trials, geographical distribution, administration routes, indications, and targets. The analysis indicates a rising prevalence of antibody-drug conjugates (ADCs), bispecific antibodies, and Fc-fusion proteins. This study will help develop new ideas for future research on antibodies in gynecology.
Collapse
Affiliation(s)
- Xiaoling Zhou
- Department of Pharmacy, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Xiao
- Department of Medical Affairs, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Fan Lai
- Department of Obstetrics, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Chen
- Department of Traditional Chinese Medicine, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Congrong Zhou
- Department of Traditional Chinese Medicine, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Deng
- Department of Pharmacy, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tao Wang
- Department of Obstetrics, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shasha Xing
- Department of Good Clinical Practice, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Haoyang Diao
- Department of Good Clinical Practice, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Mi Tang
- Department of Good Clinical Practice, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenmei Guo
- Department of Good Clinical Practice, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Erdan Luo
- Department of Good Clinical Practice, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
20
|
Wang Y, Zhang G, Rong P, Guo P, Huang S, Hang Y, Wang P, Tang L, Li X, Tang X, Ding S, Huang X, Liu J, Sun L. Intrinsic/proximal cell surface marker logic-gated extracellular targeted protein degradation in specific cell population. Mol Ther 2025:S1525-0016(25)00371-5. [PMID: 40340249 DOI: 10.1016/j.ymthe.2025.05.002] [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: 04/14/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025] Open
Abstract
Molecular tether-mediated extracellular targeted protein degradation (eTPD) presents an innovative technology and underlies a promising drug modality. However, to precisely implement eTPD within specific cell compartments remains a significant challenge. As eTPD depends on the degrader molecule expression and activity, we first seek to expand the panel of potential eTPD degraders. To this end, more than 50 receptors with variable tissue distributions are screened for identification of those with substantial endocytic rates. We subsequently assemble the bispecific, "Selected endocytic carrier-targeting chimeras (SecTAC)," and validate their efficacies to program the target cells to internalize membrane/extracellular protein cargos (or nucleic acids). Moreover, administration of a SecTAC for removal of excessive immunoglobulin G via a currently validated, emerging degrader (CD71) leads to evident therapeutic effect in a mouse lupus model. To further enhance cell-targeting specificity, we next develop logic-gated eTPD (LOG-eTPD) based on a combination of chimeras that indirectly couple cargo and degrader via another cell surface gating marker. Particularly, we find that a selective surface marker from the neighboring cells also may be exploited as input for LOG-eTPD in a therapeutically relevant context. Taken together, the present work has laid a strong foundation for developing eTPD agents that combine high potency with precision and safety.
Collapse
Affiliation(s)
- Yafeng Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Guiquan Zhang
- The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou 213000, China; College of Life Sciences, Nanjing Medical University, Changzhou 213000, China
| | - Ping Rong
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center at Medical School of Nanjing University, Nanjing 210061, China
| | - Panpan Guo
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Shisheng Huang
- The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou 213000, China; College of Life Sciences, Nanjing Medical University, Changzhou 213000, China
| | - Yang Hang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Pei Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center at Medical School of Nanjing University, Nanjing 210061, China
| | - Lin Tang
- Department of Rheumatology and Immunology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Xiaojing Li
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Shuai Ding
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Xingxu Huang
- The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou 213000, China; Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; College of Life Sciences, Nanjing Medical University, Changzhou 213000, China
| | - Jianghuai Liu
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center at Medical School of Nanjing University, Nanjing 210061, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China.
| |
Collapse
|
21
|
Cartwright D, Kidd AC, Ansel S, Ascierto ML, Spiliopoulou P. Oncogenic Signalling Pathways in Cancer Immunotherapy: Leader or Follower in This Delicate Dance? Int J Mol Sci 2025; 26:4393. [PMID: 40362630 PMCID: PMC12072740 DOI: 10.3390/ijms26094393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/01/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Immune checkpoint inhibitors have become a mainstay of treatment in many solid organ malignancies. Alongside this has been the rapid development in the identification and targeting of oncogenic drivers. The presence of alterations in oncogenic drivers not only predicts response to target therapy but can modulate the immune microenvironment and influence response to immunotherapy. Combining immune checkpoint inhibitors with targeted agents is an attractive therapeutic option but overlapping toxicity profiles may limit the clinical use of some combinations. In addition, there is growing evidence of shared resistance mechanisms that alter the response to immunotherapy when it is used after targeted therapy. Understanding this complex interaction between oncogenic drivers, targeted therapy and response to immune checkpoint inhibitors is vital for selecting the right treatment, at the right time for the right patient. In this review, we summarise the preclinical and clinical evidence of the influence of four common oncogenic alterations on immune checkpoint inhibitor response, combination therapies, and the presence of shared resistance mechanisms. We highlight the common resistance mechanisms and the need for more randomised trials investigating both combination and sequential therapy.
Collapse
Affiliation(s)
- Douglas Cartwright
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK; (D.C.); (A.C.K.); (S.A.); (M.L.A.)
- Beatson West of Scotland Cancer Centre,1053 Great Western Road, Glasgow G12 0YN, UK
| | - Andrew C. Kidd
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK; (D.C.); (A.C.K.); (S.A.); (M.L.A.)
- Beatson West of Scotland Cancer Centre,1053 Great Western Road, Glasgow G12 0YN, UK
| | - Sonam Ansel
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK; (D.C.); (A.C.K.); (S.A.); (M.L.A.)
- Beatson West of Scotland Cancer Centre,1053 Great Western Road, Glasgow G12 0YN, UK
| | - Maria Libera Ascierto
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK; (D.C.); (A.C.K.); (S.A.); (M.L.A.)
| | - Pavlina Spiliopoulou
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK; (D.C.); (A.C.K.); (S.A.); (M.L.A.)
- Beatson West of Scotland Cancer Centre,1053 Great Western Road, Glasgow G12 0YN, UK
| |
Collapse
|
22
|
Zhang J, Chang X, Bai Y, Ge X, Yin K, Xin Q. Enhanced cytotoxicity of T-DM1 in HER2-low carcinomas via autophagy inhibition. PLoS One 2025; 20:e0322029. [PMID: 40315224 PMCID: PMC12047817 DOI: 10.1371/journal.pone.0322029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/15/2025] [Indexed: 05/04/2025] Open
Abstract
Ado-trastuzumab emtansine (T-DM1), a conjugate of trastuzumab and the cytotoxic agent emtansine, has demonstrated significant antitumor efficacy in HER2-positive (HER2+) carcinoma. However, its effectiveness is limited against carcinoma cells with low HER2 expression (HER2-low). Here, we demonstrate that targeting autophagy enhances the cytotoxicity of T-DM1 against HER2-low SGC7901 cells, highlighting the potential of autophagy modulation in improving T-DM1-based therapies for HER2-low carcinomas. Specifically, this study shows that T-DM1 exhibits limited cytotoxic effects on SGC7901 cells, but pharmacological inhibition of autophagy enhances its cytotoxicity. Moreover, transmission electron microscopy revealed that autophagy activation involved the three key phases of autophagic flux: the formation, fusion, and degradation of autophagosomes, while immunoblot analysis confirmed a reduction in Akt/mTOR signaling. Furthermore, autophagy inhibition accelerated the fusion of T-DM1 with lysosomes in SGC7901 cells, as shown by confocal microscopy. Collectively, these findings suggest that while T-DM1 alone induces limited cytotoxicity, combining it with autophagy inhibitors enhances its efficacy against HER2-low carcinoma cells. Mechanistically, autophagy inhibition increases the binding of T-DM1 to lysosomes, potentially facilitating the release of emtansine from the conjugate. These results present a novel strategy that combines T-DM1 with autophagy inhibitors to effectively treat HER2-low gastric cancer, thereby broadening the therapeutic scope of T-DM1 to encompass previously challenging cancer types.
Collapse
Affiliation(s)
- Jinghui Zhang
- Department of General Surgery, The 971 Hospital of Chinese People’s Liberation Army Navy, Qingdao, P.R. China
| | - Xusheng Chang
- Department of Gastrointestinal Surgery, Changhai Hospital, Second Military Medical University, Shanghai, P. R. China.
| | - Yingcheng Bai
- Department of General Surgery, The 971 Hospital of Chinese People’s Liberation Army Navy, Qingdao, P.R. China
| | - Xiancai Ge
- Department of General Surgery, The 971 Hospital of Chinese People’s Liberation Army Navy, Qingdao, P.R. China
| | - Kai Yin
- Department of Gastrointestinal Surgery, Changhai Hospital, Second Military Medical University, Shanghai, P. R. China.
| | - Qun Xin
- Department of General Surgery, The 971 Hospital of Chinese People’s Liberation Army Navy, Qingdao, P.R. China
| |
Collapse
|
23
|
Bang I, Hattori T, Leloup N, Corrado A, Nyamaa A, Koide A, Geles K, Buck E, Koide S. Selective targeting of oncogenic hotspot mutations of the HER2 extracellular domain. Nat Chem Biol 2025; 21:706-715. [PMID: 39438724 DOI: 10.1038/s41589-024-01751-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024]
Abstract
Oncogenic mutations in the extracellular domain (ECD) of cell-surface receptors could serve as tumor-specific antigens that are accessible to antibody therapeutics. Such mutations have been identified in receptor tyrosine kinases including HER2. However, it is challenging to selectively target a point mutant, while sparing the wild-type protein. Here we developed antibodies selective to HER2 S310F and S310Y, the two most common oncogenic mutations in the HER2 ECD, via combinatorial library screening and structure-guided design. Cryogenic-electron microscopy structures of the HER2 S310F homodimer and an antibody bound to HER2 S310F revealed that these antibodies recognize the mutations in a manner that mimics the dimerization arm of HER2 and thus inhibit HER2 dimerization. These antibodies as T cell engagers selectively killed a HER2 S310F-driven cancer cell line in vitro, and in vivo as a xenograft. These results validate HER2 ECD mutations as actionable therapeutic targets and offer promising candidates toward clinical development.
Collapse
Affiliation(s)
- Injin Bang
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Takamitsu Hattori
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Nadia Leloup
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Alexis Corrado
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Atekana Nyamaa
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Akiko Koide
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Division of Hematology Oncology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Ken Geles
- Black Diamond Therapeutics, New York, NY, USA
| | | | - Shohei Koide
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA.
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
24
|
Wang R, Hu B, Pan Z, Mo C, Zhao X, Liu G, Hou P, Cui Q, Xu Z, Wang W, Yu Z, Zhao L, He M, Wang Y, Fu C, Wei M, Yu L. Antibody-Drug Conjugates (ADCs): current and future biopharmaceuticals. J Hematol Oncol 2025; 18:51. [PMID: 40307936 PMCID: PMC12044742 DOI: 10.1186/s13045-025-01704-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 04/13/2025] [Indexed: 05/02/2025] Open
Abstract
Antibody-drug conjugates (ADCs) represent a novel class of biopharmaceuticals comprising monoclonal antibodies covalently conjugated to cytotoxic agents via engineered chemical linkers. This combination enables targeted delivery of cytotoxic agents to tumor site through recognizing target antigens by antibody while minimizing off-target effects on healthy tissues. Clinically, ADCs overcome the limitations of traditional chemotherapy, which lacks target specificity, and enhance the therapeutic efficacy of monoclonal antibodies, providing higher efficacy and fewer toxicity anti-tumor biopharmaceuticals. ADCs have ushered in a new era of targeted cancer therapy, with 15 drugs currently approved for clinical use. Additionally, ADCs are being investigated as potential therapeutic candidates for autoimmune diseases, persistent bacterial infections, and other challenging indications. Despite their therapeutic benefits, the development and application of ADCs face significant challenges, including antibody immunogenicity, linker instability, and inadequate control over the release of cytotoxic agent. How can ADCs be designed to be safer and more efficient? What is the future development direction of ADCs? This review provides a comprehensive overview of ADCs, summarizing the structural and functional characteristics of the three core components, antibody, linker, and payload. Furthermore, we systematically assess the advancements and challenges associated with the 15 approved ADCs in cancer therapy, while also exploring the future directions and ongoing challenges. We hope that this work will provide valuable insights into the design and optimization of next-generation ADCs for wider clinical applications.
Collapse
Grants
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
Collapse
Affiliation(s)
- Ruili Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Baohui Hu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Ziyu Pan
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Chongxia Mo
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Xin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Guojia Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Ping Hou
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Qi Cui
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Zhao Xu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Wenjia Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
- Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, 110122, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, China
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, China
| | - Yan Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
- Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, 110122, China
| | - Chen Fu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
- Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, 110122, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
- Liaoning Medical Diagnosis and Treatment Center, Shenyang, 110000, China.
| | - Lifeng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
- Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
25
|
Dutta K, Pal D, Li S, Shyam C, Shoghi KI. Corr-A-Net: Interpretable Attention-Based Correlated Feature Learning framework for predicting of HER2 Score in Breast Cancer from H&E Images. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.22.25326227. [PMID: 40313277 PMCID: PMC12045401 DOI: 10.1101/2025.04.22.25326227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2) expression is a critical biomarker for assessing breast cancer (BC) severity and guiding targeted anti-HER2 therapies. The standard method for measuring HER2 expression is manual assessment of IHC slides by pathologists, which is both time intensive and prone to inter- and intra-observer variability. To address these challenges, we developed an interpretable deep-learning pipeline with Correlational Attention Neural Network (Corr-A-Net) to predict HER2 score from H&E images. Each prediction was accompanied with a confidence score generated by the surrogate confidence score estimation network trained using incentivized mechanism. The shared correlated representations generated using the attention mechanism of Corr-A-Net achieved the best predictive accuracy of 0.93 and AUC-ROC of 0.98. Additionally, correlated representations demonstrated the highest mean effective confidence (MEC) score of 0.85 indicating robust confidence level estimation for prediction. The Corr-A-Net can have profound implications in facilitating prediction of HER2 status from H&E images.
Collapse
Affiliation(s)
- Kaushik Dutta
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Debojyoti Pal
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Suya Li
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Chandresh Shyam
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Kooresh I. Shoghi
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
- Department of Biomedical Engineering, Washington University in St Louis, St Louis, MO USA
- Lead contact
| |
Collapse
|
26
|
Zhao X, Li Y, Zhang H, Cai Y, Wang X, Liu Y, Li T, Xu C, Teng Y, Li D, Li F. PAK5 promotes the trastuzumab resistance by increasing HER2 nuclear accumulation in HER2-positive breast cancer. Cell Death Dis 2025; 16:323. [PMID: 40258843 PMCID: PMC12012021 DOI: 10.1038/s41419-025-07657-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/23/2025]
Abstract
Nuclear HER2 (N-HER2) predicts resistance to HER2-targeted therapy and poor prognosis of breast cancer patients, and the underlying mechanisms remain unclear. Here, we show that high expression of p21-activated kinase 5 (PAK5) is associated with HER2-targeted therapy resistance and poor outcomes of breast cancer patients. Excitingly, we find an increase in N-HER2 protein expression in patients with high PAK5 expression, who demonstrate resistance to trastuzumab treatment. PAK5 phosphorylates methyltransferase METTL14 on serine 399 to enhance m6A modification of lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), leading to increased MALAT1 stability. The stabilized MALAT1 inhibits ubiquitin-proteasomal degradation of the N-HER2 by affecting the interaction of deubiquitinase USP8 and N-HER2, thereby promoting N-HER2 accumulation. Moreover, HER2 upregulates the expression of PAK5 and MALAT1, activating the HER2-MALAT1 positive feedback loop. Importantly, PAK5 promotes the therapeutic resistance of HER2-positive breast cancer cells by increasing N-HER2 protein both in vitro and vivo. These findings highlight PAK5 as a therapeutic target for combating trastuzumab resistance in HER2-positive breast cancer.
Collapse
MESH Headings
- Humans
- p21-Activated Kinases/metabolism
- p21-Activated Kinases/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Trastuzumab/pharmacology
- Trastuzumab/therapeutic use
- Female
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- RNA, Long Noncoding/metabolism
- RNA, Long Noncoding/genetics
- Animals
- Cell Line, Tumor
- Mice
- Mice, Nude
- Gene Expression Regulation, Neoplastic/drug effects
- Cell Nucleus/metabolism
- Cell Nucleus/drug effects
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Xin Zhao
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
- Department of Pediatric Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China.
| | - Hongyan Zhang
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Yihang Cai
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Xu Wang
- Department of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yidu Liu
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Tingting Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Chendong Xu
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Yuee Teng
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.
| | - Danni Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
27
|
Sarsenbayeva A, Sadak S, Kucuk I, Kudreyeva L, Bakytzhanovna AM, Uslu B. Molybdenum-Based Electrochemical Sensors for Breast Cancer Biomarker Detection: Advances and Challenges. Crit Rev Anal Chem 2025:1-21. [PMID: 40257753 DOI: 10.1080/10408347.2025.2487581] [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: 04/22/2025]
Abstract
Breast cancer, which is considered the most common type of cancer among women worldwide, is estimated to reach 4.4 million cases in 2070. Early diagnosis has become very important to prevent this expected increase. Various traditional methods, such as mammography, biopsy, enzyme immunoassay (EI), liquid biopsy, immunohistochemistry (IGH), fluorescence in situ hybridization (FISH) are used to diagnose breast cancer, but the fact that these methods are very expensive, have low sensitivity, and cause mutations in tissues due to X-rays has led researchers to discover faster, more cost-effective, and easily detectable methods. In particular, increased levels of new blood-based biomarkers in the circulation can be detected sensitively and selectively by electrochemical methods to facilitate early disease screening and rapid diagnosis. This comprehensive review focuses on the prevalence and pathology of breast cancer, clinical diagnosis of breast cancer, and electrochemical sensors of molybdenum-based compounds for the detection of various breast cancer biomarkers in recent years. Electrochemical analysis studies carried out in the field in recent years are compiled and are considered as aptamer-based, nucleotide-based, and immunosensors. The chemical properties of molybdenum compounds are discussed, and the modifications of these compounds to the electrode surface are discussed under 4 headings: drop casting, electrodeposition, atomic layer deposition, and electrophoretic deposition.
Collapse
Affiliation(s)
- Aliya Sarsenbayeva
- Department of Analytical Chemistry, Colloidal Chemistry and Technology of Rare Elements, Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Selenay Sadak
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
- The Graduate School of Health Sciences, Ankara University, Ankara, Turkey
| | - Ipek Kucuk
- The Graduate School of Health Sciences, Ankara University, Ankara, Turkey
- Department of Analytical Chemistry, Faculty of Pharmacy, Başkent University, Ankara, Turkey
| | - Leila Kudreyeva
- Department of Analytical Chemistry, Colloidal Chemistry and Technology of Rare Elements, Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Abu Moldir Bakytzhanovna
- Department of Analytical Chemistry, Colloidal Chemistry and Technology of Rare Elements, Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Bengi Uslu
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
28
|
He WZ, Yang YZ, Yin CX, Xian XY, Gu JM, Yi JH, Xue J, Zhao Y, Wang F, Hu WM, Xia LP. Evolution of HER2-low expression from primary to paired metastatic gastric cancer lesions. NPJ Precis Oncol 2025; 9:108. [PMID: 40234621 PMCID: PMC12000306 DOI: 10.1038/s41698-025-00881-1] [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: 11/04/2024] [Accepted: 03/15/2025] [Indexed: 04/17/2025] Open
Abstract
HER2-low expression has recently gained considerable attention as an actionable biomarker in gastric cancer. However, changes in HER2-low expression between primary and metastatic gastric cancers remain inadequately explored. This study included consecutive patients diagnosed with metastatic gastric cancer with both primary and metastatic tumors, between January 2014 and December 2023. HER2 status was evaluated in both primary and matched metastatic tumors. A total of 332 patients were enrolled, with HER2-negative, HER2-low, and HER2-positive statuses were observed in 226, 81, and 25 primary tumors, respectively, and in 175, 104, and 53 metastatic tumors, respectively. Among the 226 patients with HER2-negative primary tumors, 74 and 23 developed HER2-low and HER2-positive metastatic tumors, respectively. Conversion from HER2-negative primary to HER2-low metastatic gastric cancer was associated with metachronous and non-peritoneal metastasis. Overall, re-biopsy to evaluate HER2 status may be necessary, potentially broadening the patient population eligible for targeted HER2 therapy.
Collapse
Affiliation(s)
- Wen-Zhuo He
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Yuan-Zhong Yang
- Department of Pathology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Chen-Xi Yin
- Department of Intensive Care Unit, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Xin-Yi Xian
- Department of Pathology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Jia-Mei Gu
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Jia-Hong Yi
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Ju Xue
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Yue Zhao
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Fang Wang
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China.
| | - Wan-Ming Hu
- Department of Pathology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China.
| | - Liang-Ping Xia
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China.
| |
Collapse
|
29
|
He WZ, Yang YZ, Yin CX, Xian XY, Gu JM, Yi JH, Xue J, Zhao Y, Wang F, Hu WM, Xia LP. Evolution of HER2-low expression from primary to paired metastatic gastric cancer lesions. NPJ Precis Oncol 2025; 9:108. [DOI: 40234621 10.1038/s41698-025-00881-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/15/2025] [Indexed: 05/20/2025] Open
|
30
|
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
|
31
|
Justiz-Vaillant A, Pandit BR, Unakal C, Vuma S, Akpaka PE. A Comprehensive Review About the Use of Monoclonal Antibodies in Cancer Therapy. Antibodies (Basel) 2025; 14:35. [PMID: 40265416 PMCID: PMC12015915 DOI: 10.3390/antib14020035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/14/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
Monoclonal antibodies (mAbs) targeting various pathways in cancer therapy play crucial roles in enhancing the immune system's ability to recognise and eliminate tumour cells. These therapies are designed to either block inhibitory immune checkpoint pathways or to target specific tumour cell markers for direct destruction. Additionally, mAbs can modulate the tumour microenvironment, enhance antibody-dependent cellular cytotoxicity, and inhibit angiogenesis, further amplifying their therapeutic impact. Below is a summary of monoclonal antibodies targeting key pathways, along with their indications and mechanisms of action, which are reviewed based on therapeutic mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Patrick Eberechi Akpaka
- Department of Pathology/Microbiology & Pharmacology, The University of the West Indies, St. Augustine Campus, St. Augustine 330912, Trinidad and Tobago; (A.J.-V.); (B.R.P.); (C.U.); (S.V.)
| |
Collapse
|
32
|
Chintalaramulu N, Singh DP, Sapkota B, Raman D, Alahari S, Francis J. Caveolin-1: an ambiguous entity in breast cancer. Mol Cancer 2025; 24:109. [PMID: 40197489 PMCID: PMC11974173 DOI: 10.1186/s12943-025-02297-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed cancer in women and the second leading cause of death from cancer among women. Metastasis is the major cause of BC-associated mortality. Accumulating evidence implicates Caveolin-1 (Cav-1), a structural protein of plasma membrane caveolae, in BC metastasis. Cav-1 exhibits a dual role, as both a tumor suppressor and promoter depending on the cellular context and BC subtype. This review highlights the role of Cav-1 in modulating glycolytic metabolism, tumor-stromal interactions, apoptosis, and senescence. Additionally, stromal Cav-1's expression is identified as a potential prognostic marker, offering insights into its contrasting roles in tumor suppression and progression. Furthermore, Cav-1's context-dependent effects are explored in BC subtypes including hormone receptor-positive, HER2-positive, and triple-negative BC (TNBC). The review further delves into the role of Cav-1 in regulating the metastatic cascade including extracellular matrix interactions, cell migration and invasion, and premetastatic niche formation. The later sections discuss the therapeutic targeting of Cav-1 by metabolic inhibitors such as betulinic acid and Cav-1 modulating compounds. While Cav-1 may be a potential biomarker and therapeutic target, its heterogeneous expression and context-specific activity necessitates further research to develop precise interventions. Future studies investigating the mechanistic role of Cav-1 in metastasis may pave the way for effective treatment of metastatic BC.
Collapse
Affiliation(s)
- Naveen Chintalaramulu
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | | | - Biplov Sapkota
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Dayanidhi Raman
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, USA
| | | | - Joseph Francis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
33
|
Yin Q, Zhang Y, Xie X, Hou M, Chen X, Ding J. Navigating the future of gastric cancer treatment: a review on the impact of antibody-drug conjugates. Cell Death Discov 2025; 11:144. [PMID: 40188055 PMCID: PMC11972320 DOI: 10.1038/s41420-025-02429-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Gastric cancer, marked by its high incidence and poor prognosis, demands the urgent development of novel and effective treatment strategies, especially for patients ineligible for surgery or those who have had limited success with chemotherapy, radiotherapy and targeted therapies. Recently, antibody-drug conjugates (ADCs) have become a key area of investigation due to their high specificity and potent antitumor effects. These therapies combine monoclonal antibodies, designed to bind to tumor-specific antigens, with cytotoxic agents that selectively target and destroy malignant cells. ADCs have generated significant interest in clinical trials as a promising approach to improve both treatment efficacy and patient outcomes in gastric cancer. However, their clinical application is not without challenges and limitations that must be addressed. This review discusses the recent progress in the use of ADCs for gastric cancer treatment.
Collapse
Affiliation(s)
- Qingling Yin
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Yanlong Zhang
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Xueqing Xie
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Meijun Hou
- Graduate School, Zunyi Medical University, Zunyi, Guizhou, 563006, China
| | - Xunsheng Chen
- Department of Gastrointestinal Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, Guiyang, China
| | - Jie Ding
- Department of Gastrointestinal Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, Guiyang, China.
| |
Collapse
|
34
|
Mao S, Li J, Huang J, Lv L, Zhang Q, Cheng Q, Liu X, Bi Z, Yao J. Therapeutic potential of microRNA-506 in cancer treatment: mechanisms and therapeutic implications. Front Oncol 2025; 15:1524763. [PMID: 40248198 PMCID: PMC12003368 DOI: 10.3389/fonc.2025.1524763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Cancer is a complex and highly lethal disease marked by unchecked cell proliferation, aggressive behavior, and a strong tendency to metastasize. Despite significant advancements in cancer diagnosis and treatment, challenges such as early detection difficulties, drug resistance, and adverse effects of radiotherapy or chemotherapy continue to threaten patient survival. MicroRNAs (miRNAs) have emerged as critical regulators in cancer biology, with miR-506 being extensively studied and recognized for its tumor-suppressive effects across multiple cancer types. This review examines the regulatory mechanisms of miR-506 in common cancers, focusing on its role in the competing endogenous RNA (ceRNA) network and its effects on cancer cell proliferation, apoptosis, and migration. We also discuss the potential of miR-506 as a therapeutic target and its role in overcoming drug resistance in cancer treatment. Overall, these insights underscore the therapeutic potential of miR-506 and its promise in developing novel cancer therapies.
Collapse
Affiliation(s)
- Shuzhen Mao
- Department of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Junyan Li
- Department of Pathology, Second People’s Hospital of Ningyang, Taian, Shandong, China
| | - Jiahui Huang
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Lili Lv
- Department of Pathology, Second People’s Hospital of Ningyang, Taian, Shandong, China
| | - Qilian Zhang
- Department of Pathology, People’s Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qing Cheng
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Xiaojing Liu
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Zhiwei Bi
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Jing Yao
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
35
|
Li Z, Wang Y, Sun Y, Wang L, Li X, Sun L, He Z, Yang H, Wang Y, Wang Q, Song Z, Hong W, Wang Y, Xia G, Yu Y, Peng M, Song Y, Wang D, Meng R, Fang J, Luo Y, Liang W, Hu S, Wang Z, Song K, Li Y, Yang L, Shi W, Lu S. Trastuzumab rezetecan, a HER2-directed antibody-drug conjugate, in patients with advanced HER2-mutant non-small-cell lung cancer (HORIZON-Lung): phase 2 results from a multicentre, single-arm study. Lancet Oncol 2025; 26:437-446. [PMID: 40020696 DOI: 10.1016/s1470-2045(25)00012-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/19/2024] [Accepted: 01/14/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Trastuzumab rezetecan (also known as SHR-A1811) is a novel antibody-drug conjugate consisting of a humanised HER2-directed monoclonal antibody, cleavable tetrapeptide-based linker, and DNA topoisomerase I inhibitor. In the phase 1 portion of this phase 1/2 study, trastuzumab rezetecan showed preliminary anti-tumour activity and a favourable safety profile in patients with HER2-mutant non-small-cell lung cancer (NSCLC). We present phase 2 results from the study, which aimed to further evaluate the activity and safety of trastuzumab rezetecan at the recommended dose. METHODS In this multicentre, single-arm, phase 2 trial, conducted in 35 hospitals in China, we recruited patients aged 18-75 years, with locally advanced or metastatic NSCLC with an activating HER2 mutation and an Eastern Cooperative Oncology Group performance status score of 0-1, who had disease progression after or were intolerant to platinum-based chemotherapy and anti-PD-1 treatment or anti-PD-L1 treatment. Trastuzumab rezetecan was administered at 4·8 mg/kg intravenously once every 3 weeks. The primary endpoint was objective response rate assessed by an independent review committee in patients who received at least one cycle of study treatment. All patients who received at least one cycle of study treatment were included in safety analyses. This study is registered with ClinicalTrials.gov, NCT04818333, and is ongoing but not recruiting. FINDINGS Between April 14, 2023, and Dec 14, 2023, 94 patients were enrolled and treated. 42 (45%) patients were male, 52 (55%) female, 92 (98%) were Han Chinese, and two (2%) were other ethnicity Chinese. At data cutoff (June 14, 2024), the median duration of follow-up was 8·7 months (IQR 7·0-10·4). 69 (73%; 95% CI 63·3-82·0) of 94 patients had a confirmed objective response, as assessed by independent review committee. The most common grade 3-4 treatment-related adverse events were decreased neutrophil count (38 [40%] patients), decreased white blood cell count (25 [27%]), anaemia (22 [23%]), decreased platelet count (10 [11%]), and decreased lymphocyte count (seven [7%]). Treatment-related serious adverse events occurred in 22 (23%) patients, which were decreased platelet count (six [6%]), decreased neutrophil count (six [6%]), interstitial lung disease (five [5%]), decreased white blood cell count (four [4%]), anaemia (four [4%]), vomiting (three [3%]), pneumonia (three [3%]), hyponatraemia (two [2%]), and pyrexia (one [1%]), small intestinal obstruction (one [1%]), nausea (one [1%]), and chronic obstructive pulmonary disease (one [1%]). There were no treatment-related deaths. INTERPRETATION Trastuzumab rezetecan showed clinically meaningful activity and manageable safety in patients with previously treated HER2-mutant NSCLC. Further trials are justified. FUNDING Jiangsu Hengrui Pharmaceuticals, National Multi-disciplinary Treatment Project for Major Diseases, Collaborative Innovation Center for Clinical and Translational Science by the Ministry of Education & Shanghai. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Female
- Humans
- Male
- Middle Aged
- Young Adult
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Camptothecin/administration & dosage
- Camptothecin/adverse effects
- Camptothecin/analogs & derivatives
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/mortality
- China
- Immunoconjugates/administration & dosage
- Immunoconjugates/adverse effects
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Lung Neoplasms/mortality
- Mutation
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/antagonists & inhibitors
- Trastuzumab/administration & dosage
- Trastuzumab/adverse effects
- Topoisomerase I Inhibitors/administration & dosage
- Topoisomerase I Inhibitors/adverse effects
Collapse
Affiliation(s)
- Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Wang
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuping Sun
- Phase I Clinical Research Center, The Affiliated Cancer Hospital of Shandong First Medical University, Jinan, China
| | - Linlin Wang
- Chest Radiotherapy Third Ward, The Affiliated Cancer Hospital of Shandong First Medical University, Jinan, China
| | - Xingya Li
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Longhua Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhiyi He
- Pulmonary and Critical Care Medicine Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Haiyan Yang
- Department of Lung & Gastrointestinal Oncology, Hunan Cancer Hospital & The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University-Henan Cancer Hospital & Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, China
| | - Zhengbo Song
- Phase I Clinical Trial Ward, Zhejiang Cancer Hospital, Hangzhou, China
| | - Wei Hong
- Department of Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yong Wang
- Department of Medical Oncology, Anhui Provincial Hospital-The First Affiliated Hospital of the University of Science and Technology of China, Hefei, China
| | - Guohao Xia
- Department of Medical Oncology, Jiangsu Cancer Hospital-Jiangsu Institute of Cancer Research-The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Yu
- Department of Thoracic Medicine, Harbin Medical University Cancer Hospital, Harbin, China
| | - Min Peng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Donglin Wang
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Rui Meng
- Oncology Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Fang
- Department of Thoracic Oncology, Peking University Cancer Hospital, Beijing, China
| | - Yongzhong Luo
- Department of Thoracic Medicine, Hunan Cancer Hospital & The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Sheng Hu
- Department of Thoracic Oncology, Hubei Cancer Hospital, Wuhan, China
| | - Zhihui Wang
- Department of Thoracic Oncology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Ke Song
- Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | - You Li
- Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | - Lulu Yang
- Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | - Wei Shi
- Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
36
|
Liu X, Fan X, Gao X, Hu W, Sun P. Leveraging HER2-targeted biparatopic antibodies in solid tumors. Pharmacol Res 2025; 214:107687. [PMID: 40054541 DOI: 10.1016/j.phrs.2025.107687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/14/2025] [Accepted: 03/04/2025] [Indexed: 03/23/2025]
Abstract
Biparatopic antibodies (bpAbs), which target non-overlapping epitopes on the same antigen, offer unique mechanisms of action and therapeutic applications that surpass those of conventional monospecific antibodies. These distinctive properties have positioned bpAbs as effective therapeutic agents in the treatment of cancer and infectious diseases, especially in cases where current treatments face limitations. Among these, HER2-targeted bpAbs have shown significant improvements in survival outcomes for patients with solid tumors that depend on HER2 signaling. However, a comprehensive understanding of their clinical impact, mechanisms of action, and limitations in therapeutic use remains lacking. Here, we review and contrast the clinical performance of the well-established HER2-targeted bpAbs in current use, with a focus on their mechanisms of action, associated limitations, and potential combination strategies. We also highlight emerging investigational bpAbs-based agents that have shown promise in the treatment of HER2-positive solid cancers. These advancements may lead to enhanced therapeutic options and potentially broaden the scope of bpAbs in cancer therapy.
Collapse
Affiliation(s)
- Xinlin Liu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao 266071, China
| | - Xinyi Fan
- Department of Allergy, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xiang Gao
- Department of Allergy, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Weiyu Hu
- Department of Hepatobiliary and pancreatic surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Peng Sun
- Department of Hepatobiliary and pancreatic surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
37
|
Salinaro J, Singh K, Sands N, Gill V, Perati S, James N, Sharma S, Nasir A, DiSilvestro P, Miller K, Oliver M, Mathews C. Distribution and concordance of HER2 scores in endometrial and ovarian cancer. Gynecol Oncol 2025; 195:115-121. [PMID: 40088505 DOI: 10.1016/j.ygyno.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
OBJECTIVES Although multiple HER2 scoring criteria exist, the optimal strategy to identify patients with gynecologic malignancies who may benefit from HER2-directed therapies remains unknown. The objectives of this study were to assess the distribution and concordance of HER2 scores in endometrial and ovarian cancer. METHODS One hundred five tumor specimens from 94 patients with endometrial or epithelial ovarian cancer (EOC) who underwent Caris tumor profiling from 11/2022 to 01/2025 were identified from a retrospective database. Each sample was assigned a HER2 immunohistochemistry (IHC) score of 0, 1+, 2+, or 3+ using breast, endometrial, and gastric criteria. ERBB2 amplification and HER2 IHC scores were abstracted from Caris reports. Patient characteristics and HER2 score distributions were analyzed using descriptive statistics and Fisher's exact test. Matrix correlation coefficients were used to assess HER2 score concordance. RESULTS A total of 105 samples underwent internal triplicate HER2 scoring - 63 EOC and 42 endometrial. A higher percentage of patients with endometrial cancer were HER2-high compared to those with EOC (45.2-50 % vs 20.6 %, p < 0.05). Internal triplicate HER2 score concordance was strong (r ≥ 0.96, p < 0.001) but weaker when compared to Caris scores (r = 0.66). Of the 23 discordant HER2 results, 13 would have changed therapy eligibility (56.5 %). Only 12 patients (12.7 %) had intermediate or high ERBB2 amplification. CONCLUSIONS A clinically significant percentage of patients had HER2-high tumors regardless of tumor type. HER2 score concordance was strong within each sample but weaker when compared to Caris scores. Incorporating multi-site testing and/or validation of external IHC into any gynecologic HER2 scoring algorithm should be considered.
Collapse
Affiliation(s)
- Julia Salinaro
- Program in Women's Oncology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA.
| | - Kamaljeet Singh
- Department of Pathology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Natalie Sands
- Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Victoria Gill
- Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Shriya Perati
- Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Nicole James
- Program in Women's Oncology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Shreenidhi Sharma
- Hassenfeld Child Health and Innovation Institute, School of Public Health, Brown University, Providence, RI, USA
| | | | - Paul DiSilvestro
- Program in Women's Oncology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Katherine Miller
- Program in Women's Oncology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Matthew Oliver
- Program in Women's Oncology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Cara Mathews
- Program in Women's Oncology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| |
Collapse
|
38
|
Shaik R, Mounika V, Begum S, Rajkumar A, Mallikarjun B, Sri Harshini V, Kolure R, Sreevani B, Thakur S. Monoclonal Antibodies in Clinical Trials for Breast Cancer Treatment. Monoclon Antib Immunodiagn Immunother 2025; 44:17-39. [PMID: 40171653 DOI: 10.1089/mab.2024.0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025] Open
Abstract
One of the most potent therapeutic and diagnostic agents in contemporary medicine is the monoclonal antibody (mAb). mAbs can perform a variety of tasks in breast cancer (BC), including identifying and delivering therapeutic medications to targets, preventing cell development, and suppressing immune system inhibitors including directly attacking cancer cells. mAbs are one of the most effective therapeutic options, particularly for HER2, but they have not been well studied for their use in treating other forms of BC, particularly triple negative breast tumors. Bispecific and trispecific mAbs have created new opportunities for more targeted specific efficacy, which has a positive impact on the viability of antigen specificity. They are more versatile and effective than other forms of treatment, emerging as most popular option for treating BC. However, mAbs have a limit in treatment due to certain adverse effects, including fever, shaking, exhaustion, headache, nausea, and vomiting, as well as rashes, bleeding, and difficulty breathing. To examine the current and prospective future capacities of mAbs with regard to the detection and treatment of BC, the present review highlights advantages and disadvantages of mAb approach.
Collapse
Affiliation(s)
- Rahaman Shaik
- School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Varikuppala Mounika
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Shireen Begum
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Agolapu Rajkumar
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Bathurasi Mallikarjun
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Vollala Sri Harshini
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Rajini Kolure
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | | | - Sneha Thakur
- Department of Pharmacognosy, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| |
Collapse
|
39
|
El Hage M, Su Z, Linnebacher M. Addressing Challenges in Targeted Therapy for Metastatic Colorectal Cancer. Cancers (Basel) 2025; 17:1098. [PMID: 40227578 PMCID: PMC11988006 DOI: 10.3390/cancers17071098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 04/15/2025] Open
Abstract
This review article aims to address the challenges associated with targeted therapy for the treatment of metastatic colorectal cancer (mCRC). We will first provide an overview of approved targeted therapies for treating mCRC, which include antiangiogenic therapy, as well as inhibitors of EGFR, BRAFV600E, HER2 inhibitors, and immune checkpoints. Second, we discuss the different mechanisms of primary resistance, including tumor heterogeneity, both as inter-patient and intra-patient heterogeneity, and mechanisms of secondary resistance which include: driver oncogene alterations, downstream or parallel bypass signaling, presence of co-dominant driver oncogenes, tumor lineage plasticity, and epithelial to mesenchymal transition. Resistance mechanisms towards the different drug classes targeting mCRC are discussed in detail. Strategies to overcome resistance primarily involve combination of therapies, although this approach is typically linked to increased drug toxicity, manifesting as on and off-target effects. Moreover, the cost and accessibility of targeted therapies pose significant challenges for diverse populations. Addressing these challenges necessitates further research efforts aimed at optimizing the use of targeted therapy in mCRC. Integration of genomic biomarkers, such as sequencing and liquid biopsy, into routine clinical practice holds promise in enhancing treatment outcomes. In conclusion, this comprehensive review underscores the complex challenges encountered in targeted therapy for mCRC.
Collapse
Affiliation(s)
| | | | - Michael Linnebacher
- Molecular Oncology and Immunotherapy, Clinic of General Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (M.E.H.); (Z.S.)
| |
Collapse
|
40
|
Singh SR, Bhaskar R, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Mladenov M, Hadzi-Petrushev N, Stojchevski R, Sinha JK, Avtanski D. Exploring the Genetic Orchestra of Cancer: The Interplay Between Oncogenes and Tumor-Suppressor Genes. Cancers (Basel) 2025; 17:1082. [PMID: 40227591 PMCID: PMC11988167 DOI: 10.3390/cancers17071082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Cancer is complex because of the critical imbalance in genetic regulation as characterized by both the overexpression of oncogenes (OGs), mainly through mutations, amplifications, and translocations, and the inactivation of tumor-suppressor genes (TSGs), which entail the preservation of genomic integrity by inducing apoptosis to counter the malignant growth. Reviewing the intricate molecular interplay between OGs and TSGs draws attention to their cell cycle, apoptosis, and cancer metabolism regulation. In the present review, we discuss seminal discoveries, such as Knudson's two-hit hypothesis, which framed the field's understanding of cancer genetics, leading to the next breakthroughs with next-generation sequencing and epigenetic profiling, revealing novel insights into OG and TSG dysregulation with opportunities for targeted therapy. The key pathways, such as MAPK/ERK, PI3K/AKT/mTOR, and Wnt/β-catenin, are presented in the context of tumor progression. Importantly, we further highlighted the advances in therapeutic strategies, including inhibitors of KRAS and MYC and restoration of TSG function, despite which mechanisms of resistance and tumor heterogeneity pose daunting challenges. A high-level understanding of interactions between OG-TSGs forms the basis for effective, personalized cancer treatment-something to strive for in better clinical outcomes. This synthesis should integrate foundational biology with translation and, in this case, contribute to the ongoing effort against cancer.
Collapse
Affiliation(s)
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan-si 38541, Republic of Korea;
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan-si 38541, Republic of Korea
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, India
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Symbiosis International (Deemed University), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, India
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
| | - Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | | | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
41
|
Ye Z, Yuan J, Hong D, Xu P, Liu W. Multimodal diagnostic models and subtype analysis for neoadjuvant therapy in breast cancer. Front Immunol 2025; 16:1559200. [PMID: 40170854 PMCID: PMC11958217 DOI: 10.3389/fimmu.2025.1559200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
Background Breast cancer, a heterogeneous malignancy, comprises multiple subtypes and poses a substantial threat to women's health globally. Neoadjuvant therapy (NAT), administered prior to surgery, is integral to breast cancer treatment strategies. It aims to downsize tumors, optimize surgical outcomes, and evaluate tumor responsiveness to treatment. However, accurately predicting NAT efficacy remains challenging due to the disease's complexity and the diverse responses across different molecular subtypes. Methods In this study, we harnessed multimodal data, including proteomic, genomic, MRI imaging, and clinical information, sourced from multiple cohorts such as I-SPY2, TCGA-BRCA, GSE161529, and METABRIC. Post data preprocessing, Lasso regression was utilized for feature extraction and selection. Five machine learning algorithms were employed to construct diagnostic models, with pathological complete response (pCR) as the predictive endpoint. Results Our results revealed that the multi-omics Ridge regression model achieved the optimal performance in predicting pCR, with an AUC of 0.917. Through unsupervised clustering using the R package MOVICS and nine clustering algorithms, we identified four distinct multimodal breast cancer subtypes associated with NAT. These subtypes exhibited significant differences in proteomic profiles, hallmark cancer gene sets, pathway activities, tumor immune microenvironments, transcription factor activities, and clinical characteristics. For instance, CS1 subtype, predominantly ER-positive, had a low pCR rate and poor response to chemotherapy drugs, while CS4 subtype, characterized by high immune infiltration, showed a better response to immunotherapy. At the single-cell level, we detected significant heterogeneity in the tumor microenvironment among the four subtypes. Malignant cells in different subtypes displayed distinct copy number variations, differentiation levels, and evolutionary trajectories. Cell-cell communication analysis further highlighted differential interaction patterns among the subtypes, with implications for tumor progression and treatment response. Conclusion Our multimodal diagnostic model and subtype analysis provide novel insights into predicting NAT efficacy in breast cancer. These findings hold promise for guiding personalized treatment strategies. Future research should focus on experimental validation, in-depth exploration of the underlying mechanisms, and extension of these methods to other cancers and treatment modalities.
Collapse
Affiliation(s)
- Zheng Ye
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, China
| | - Jiaqi Yuan
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Deqing Hong
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Peng Xu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, China
| | - Wenbin Liu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| |
Collapse
|
42
|
Wang T, He M, Guan W. Pyrotinib monotherapy for advanced HER2-positive esophageal adenocarcinoma with trastuzumab resistance and chemotherapy intolerance: a case report and literature review. Discov Oncol 2025; 16:335. [PMID: 40095240 PMCID: PMC11914708 DOI: 10.1007/s12672-025-02049-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
HER2-positive advanced esophageal adenocarcinoma (EAC) cases demonstrate a poor prognosis because of drug resistance that develops after standard first-line trastuzumab therapy. The patient was initially diagnosed with stage cT2N1M0 III EAC. He underwent neoadjuvant chemotherapy, radical esophageal resection, and postoperative adjuvant radiotherapy. However, four months after treatment, the lesion relapsed and progressed to the right back, rendering the case inoperable. Pathological analysis revealed HER2 amplification. Given a poor tolerance to chemotherapy, the patient was administered cadonilimab and trastuzumab for three months. Subsequently, the second-line therapy was switched to pyrotinib monotherapy as a salvage treatment. Remarkably, after one month of treatment, the tumor showed significant reduction, with mild toxic side effects. Pyrotinib can be used for salvage later-line therapy in HER2-positive advanced EAC with trastuzumab resistance or poor chemotherapy tolerance, which deserves further promotion.
Collapse
Affiliation(s)
- Tao Wang
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Mingyuan He
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Wei Guan
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| |
Collapse
|
43
|
Zhang Z, Wang Y, Lu W, Wang X, Guo H, Pan X, Liu Z, Wu Z, Qin W. Spatiotemporally resolved mapping of extracellular proteomes via in vivo-compatible TyroID. Nat Commun 2025; 16:2553. [PMID: 40089463 PMCID: PMC11910615 DOI: 10.1038/s41467-025-57767-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Extracellular proteins play pivotal roles in both intracellular signaling and intercellular communications in health and disease. While recent advancements in proximity labeling (PL) methods, such as peroxidase- and photocatalyst-based approaches, have facilitated the resolution of extracellular proteomes, their in vivo compatibility remains limited. Here, we report TyroID, an in vivo-compatible PL method for the unbiased mapping of extracellular proteins with high spatiotemporal resolution. TyroID employs plant- and bacteria-derived tyrosinases to produce reactive o-quinone intermediates, enabling the labeling of multiple residues on endogenous proteins with bioorthogonal handles, thereby allowing for their identification via chemical proteomics. We validate TyroID's specificity by mapping extracellular proteomes and HER2-neighboring proteins using affibody-directed recombinant tyrosinases. Demonstrating its superiority over other PL methods, TyroID enables in vivo mapping of extracellular proteomes, including mapping HER2-proximal proteins in tumor xenografts, quantifying the turnover of plasma proteins and labeling hippocampal-specific proteomes in live mouse brains. TyroID emerges as a potent tool for investigating protein localization and molecular interactions within living organisms.
Collapse
Affiliation(s)
- Zijuan Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China
- The State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
| | - Yankun Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Wenjie Lu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xiaofei Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Hongyang Guo
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xuanzhen Pan
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Zeyu Liu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Zhaofa Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Wei Qin
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China.
- The State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China.
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China.
| |
Collapse
|
44
|
Molefi T, Mabonga L, Hull R, Mwazha A, Sebitloane M, Dlamini Z. The Histomorphology to Molecular Transition: Exploring the Genomic Landscape of Poorly Differentiated Epithelial Endometrial Cancers. Cells 2025; 14:382. [PMID: 40072110 PMCID: PMC11898822 DOI: 10.3390/cells14050382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
The peremptory need to circumvent challenges associated with poorly differentiated epithelial endometrial cancers (PDEECs), also known as Type II endometrial cancers (ECs), has prompted therapeutic interrogation of the prototypically intractable and most prevalent gynecological malignancy. PDEECs account for most endometrial cancer-related mortalities due to their aggressive nature, late-stage detection, and poor response to standard therapies. PDEECs are characterized by heterogeneous histopathological features and distinct molecular profiles, and they pose significant clinical challenges due to their propensity for rapid progression. Regardless of the complexities around PDEECs, they are still being administered inefficiently in the same manner as clinically indolent and readily curable type-I ECs. Currently, there are no targeted therapies for the treatment of PDEECs. The realization of the need for new treatment options has transformed our understanding of PDEECs by enabling more precise classification based on genomic profiling. The transition from a histopathological to a molecular classification has provided critical insights into the underlying genetic and epigenetic alterations in these malignancies. This review explores the genomic landscape of PDEECs, with a focus on identifying key molecular subtypes and associated genetic mutations that are prevalent in aggressive variants. Here, we discuss how molecular classification correlates with clinical outcomes and can refine diagnostic accuracy, predict patient prognosis, and inform therapeutic strategies. Deciphering the molecular underpinnings of PDEECs has led to advances in precision oncology and protracted therapeutic remissions for patients with these untamable malignancies.
Collapse
Affiliation(s)
- Thulo Molefi
- Discipline of Obstetrics and Gynaecology, School of Clinical Medicine, University of KwaZulu-Natal, Durban 4002, South Africa;
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP) Pan African Research Institute (PACRI), University of Pretoria, Hartfield, Pretoria 0028, South Africa
- Department of Medical Oncology, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Lloyd Mabonga
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP) Pan African Research Institute (PACRI), University of Pretoria, Hartfield, Pretoria 0028, South Africa
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP) Pan African Research Institute (PACRI), University of Pretoria, Hartfield, Pretoria 0028, South Africa
| | - Absalom Mwazha
- Department of Anatomical Pathology, National Health Laboratory Services, Durban 4058, South Africa
| | - Motshedisi Sebitloane
- Discipline of Obstetrics and Gynaecology, School of Clinical Medicine, University of KwaZulu-Natal, Durban 4002, South Africa;
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP) Pan African Research Institute (PACRI), University of Pretoria, Hartfield, Pretoria 0028, South Africa
| |
Collapse
|
45
|
Gerber WK, Xie Y, Patel SA. Expanding the Therapeutic Reach of Chimeric Antigen Receptor T-Cells and Bispecific T-Cell Engagers Across Solid Tumors. JCO Precis Oncol 2025; 9:e2400753. [PMID: 40138603 PMCID: PMC11952672 DOI: 10.1200/po-24-00753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/29/2025] [Accepted: 02/06/2025] [Indexed: 03/29/2025] Open
Abstract
The introduction of T-cell-based therapeutics in hematologic malignancies has led to improvements in outcomes for patients with acute leukemia, lymphoma, and multiple myeloma. To date, the Food and Drug Administration (FDA) has approved seven chimeric antigen receptor-T (CAR-T) cell therapies and seven bispecific T-cell engagers (BiTEs) across a variety of hematologic malignancies; however, the extension of CAR-T therapies and BiTEs to the solid tumor arena has been somewhat limited. In this review, we discuss the landmark data that led to the commercialization of four novel FDA-approved T-cell-based therapeutics in solid malignancies, including tarlatamab for small cell lung cancer, afamitresgene autoleucel for synovial sarcoma, lifileucel for metastatic melanoma, and tebentafusp for metastatic uveal melanoma. We discuss the targetable antigen landscape of CAR-T therapies and BiTEs under investigation in solid malignancies. We explore the translational potential for various CARs under active investigation, including human epidermal growth factor receptor 2-directed CARs in breast cancer, prostate stem cell antigen-directed CARs for prostate cancer, epidermal growth factor receptor (EGFR)-IL13Ra2 and EGFR-vIII CARs for glioblastoma, and GD2-directed CARs for neuroblastoma. We glean from lessons learned for existing CAR-T therapies and BiTEs for hematologic malignancies and emphasize solutions toward facilitating the clinical rollout of T-cell-based therapies in solid tumors, including scalability to meet the growing needs of clinical oncology. Some solutions include addressing on-target, off-tumor toxicity; improving the manufacturing of CARs; optimizing the tissue-specific tumor microenvironment by combating immune desert tumors; and discovering natural tumor neoantigens and non-self-epitopes generated by tumor-specific mutations. These concepts can help provide transformative benefits for patients with solid malignancies in the coming years.
Collapse
Affiliation(s)
- William K. Gerber
- Dept. of Medicine – Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA
| | - Yiyu Xie
- Dept. of Medicine – Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA
| | - Shyam A. Patel
- Dept. of Medicine – Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA
- Center for Clinical and Translational Science, UMass Chan Medical School, Worcester, MA
- Cancer Biology Program, Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA
| |
Collapse
|
46
|
Landi D, Navai SA, Brock RM, Fousek K, Nawas Z, Sanber K, Chauvin-Fleurence C, Bhat RR, Xu S, Krishnamurthy P, Choe M, Campbell ME, Morris JS, Gad AZ, Shree A, Echeandia Marrero AS, Saadeldin AM, Matthew PR, Mullikin D, Bielamowicz K, Kurenbekova L, Major AM, Salsman VS, Byrd TT, Hicks JM, Zhang YJ, Yustein J, Carisey AF, Joseph SK, Ahmed N, Hegde M. A Checkpoint Reversal Receptor Mediates Bipartite Activation and Enhances CAR T-cell Function. CANCER RESEARCH COMMUNICATIONS 2025; 5:527-548. [PMID: 39973814 PMCID: PMC11955954 DOI: 10.1158/2767-9764.crc-24-0125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 09/18/2024] [Accepted: 02/17/2025] [Indexed: 02/21/2025]
Abstract
SIGNIFICANCE Enhancing CART function and persistence while balancing immune effector-mediated inflammation is crucial. Using our clinically relevant HER2-CAR platform, we demonstrate that tumor-intrinsic signals like the PD-1/PD-L1 immune checkpoint can be leveraged in CART design to modulate immune synapse and metabolic parameters, improving antitumor function without increasing cytokine production.
Collapse
Affiliation(s)
- Daniel Landi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Shoba A. Navai
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Rebecca M. Brock
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Kristen Fousek
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - Zeid Nawas
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Khaled Sanber
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Cynthia Chauvin-Fleurence
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Raksha R. Bhat
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Shuo Xu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Purna Krishnamurthy
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Michelle Choe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Matthew E. Campbell
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Jessica S. Morris
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - Ahmed Z. Gad
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - Ankita Shree
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Alesandra S. Echeandia Marrero
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Amr M. Saadeldin
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX
| | - Pretty R. Matthew
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Dolores Mullikin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Kevin Bielamowicz
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Lyazat Kurenbekova
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Angela M. Major
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Vita S. Salsman
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Tiara T. Byrd
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - John M. Hicks
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Yi Jonathan Zhang
- Department of Neurosurgery, Houston Methodist Hospital, Houston, Texas
| | - Jason Yustein
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Alexandre F. Carisey
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Cell & Molecular Biology Department, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Sujith K. Joseph
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Nabil Ahmed
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Meenakshi Hegde
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
47
|
Yin J, Zhang H, Sun X, You N, Mou M, Lu M, Pan Z, Li F, Li H, Zeng S, Zhu F. Decoding Drug Response With Structurized Gridding Map-Based Cell Representation. IEEE J Biomed Health Inform 2025; 29:1702-1713. [PMID: 38090819 DOI: 10.1109/jbhi.2023.3342280] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025]
Abstract
A thorough understanding of cell-line drug response mechanisms is crucial for drug development, repurposing, and resistance reversal. While targeted anticancer therapies have shown promise, not all cancers have well-established biomarkers to stratify drug response. Single-gene associations only explain a small fraction of the observed drug sensitivity, so a more comprehensive method is needed. However, while deep learning models have shown promise in predicting drug response in cell lines, they still face significant challenges when it comes to their application in clinical applications. Therefore, this study proposed a new strategy called DD-Response for cell-line drug response prediction. First, a limitation of narrow modeling horizons was overcome to expand the model training domain by integrating multiple datasets through source-specific label binarization. Second, a modified representation based on a two-dimensional structurized gridding map (SGM) was developed for cell lines & drugs, avoiding feature correlation neglect and potential information loss. Third, a dual-branch, multi-channel convolutional neural network-based model for pairwise response prediction was constructed, enabling accurate outcomes and improved exploration of underlying mechanisms. As a result, the DD-Response demonstrated superior performance, captured cell-line characteristic variations, and provided insights into key factors impacting cell-line drug response. In addition, DD-Response exhibited scalability in predicting clinical patient responses to drug therapy. Overall, because of DD-response's excellent ability to predict drug response and capture key molecules behind them, DD-response is expected to greatly facilitate drug discovery, repurposing, resistance reversal, and therapeutic optimization.
Collapse
|
48
|
Bitzer M, Groß S, Albert J, Blödt S, Boda-Heggemann J, Borucki K, Brunner T, Caspari R, Dombrowski F, Evert M, Follmann M, Freudenberger P, Gani C, Gebert J, Geier A, Gkika E, Götz M, Helmberger T, Hoffmann RT, Huppert P, Krug D, La Fougère C, Lang H, Langer T, Lenz P, Lüdde T, Mahnken A, Nadalin S, Nguyen HHP, Nothacker M, Ockenga J, Oldhafer K, Ott J, Paprottka P, Pereira P, Persigehl T, Plentz R, Pohl J, Recken H, Reimer P, Riemer J, Ringe K, Roeb E, Rüssel J, Schellhaas B, Schirmacher P, Schlitt HJ, Schmid I, Schütte K, Schuler A, Seehofer D, Sinn M, Stengel A, Steubesand N, Stoll C, Tannapfel A, Taubert A, Trojan J, van Thiel I, Utzig M, Vogel A, Vogl T, Wacker F, Waidmann O, Wedemeyer H, Wege H, Wenzel G, Wildner D, Wörns MA, Galle P, Malek N. [Not Available]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2025; 63:e159-e260. [PMID: 40064172 DOI: 10.1055/a-2460-6298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2025]
Affiliation(s)
- Michael Bitzer
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| | - Sabrina Groß
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| | - Jörg Albert
- Katharinenhospital, Klinik für Allgemeine Innere Medizin, Gastroenterologie, Hepatologie, Infektiologie und Pneumologie, Stuttgart
| | - Susanne Blödt
- Arbeitsgemeinschaft der Wissenschaftlichen Medizinischen Fachgesellschaften e. V. (AWMF), Berlin
| | | | - Katrin Borucki
- Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Institut für Klinische Chemie und Pathobiochemie
| | - Thomas Brunner
- Universitätsklinik für Strahlentherapie-Radioonkologie, Medizinische Universität Graz
| | - Reiner Caspari
- Klinik Niederrhein Erkrankungen des Stoffwechsels der Verdauungsorgane und Tumorerkrankungen, Bad Neuenahr-Ahrweiler
| | | | | | - Markus Follmann
- Office des Leitlinienprogrammes Onkologie, Deutsche Krebsgesellschaft e.V., Berlin
| | | | - Cihan Gani
- Klinik für Radioonkologie, Universitätsklinikum Tübingen
| | - Jamila Gebert
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| | - Andreas Geier
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg
| | - Eleni Gkika
- Klinik für Strahlenheilkunde, Department für Radiologische Diagnostik und Therapie, Universitätsklinikum Freiburg
| | - Martin Götz
- Medizinische Klinik IV - Gastroenterologie/Onkologie, Klinikverbund Südwest, Böblingen
| | - Thomas Helmberger
- Institut für Radiologie, Neuroradiologie und minimal invasive Therapie, München Klinik Bogenhausen
| | - Ralf-Thorsten Hoffmann
- Institut und Poliklinik für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Dresden
| | - Peter Huppert
- Radiologisches Zentrum, Max Grundig Klinik, Bühlerhöhe
| | - David Krug
- Strahlentherapie Campus Kiel, Universitätsklinikum Schleswig-Holstein
| | - Christian La Fougère
- Nuklearmedizin und Klinische Molekulare Bildgebung, Eberhard-Karls Universität, Tübingen
| | - Hauke Lang
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Johannes Gutenberg-Universität, Mainz
| | - Thomas Langer
- Office des Leitlinienprogrammes Onkologie, Deutsche Krebsgesellschaft e.V., Berlin
| | - Philipp Lenz
- Zentrale Einrichtung Palliativmedizin, Universitätsklinikum Münster
| | - Tom Lüdde
- Medizinische Klinik für Gastroenterologie, Hepatologie und Infektiologie, Universitätsklinikum Düsseldorf
| | - Andreas Mahnken
- Klinik für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Marburg
| | - Silvio Nadalin
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Eberhard-Karls Universität, Tübingen
| | | | - Monika Nothacker
- Arbeitsgemeinschaft der Wissenschaftlichen Medizinischen Fachgesellschaften e. V. (AWMF), Berlin
| | - Johann Ockenga
- Medizinische Klinik II, Gesundheit Nord, Klinikverbund Bremen
| | - Karl Oldhafer
- Klinik für Leber-, Gallenwegs- und Pankreaschirurgie, Asklepios Klinik Barmbek
| | - Julia Ott
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| | - Philipp Paprottka
- Sektion für Interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München
| | - Philippe Pereira
- Zentrum für Radiologie, Minimal-invasive Therapien und Nuklearmedizin, SLK-Klinken Heilbronn
| | - Thorsten Persigehl
- Institut für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Köln
| | - Ruben Plentz
- Digestive Diseases and Nutrition, Gastroenterology, University of Kentucky
| | - Jürgen Pohl
- Abteilung für Gastroenterologie, Asklepios Klinik Altona
| | | | - Peter Reimer
- Institut für Diagnostische und Interventionelle Radiologie, Städtisches Klinikum Karlsruhe
| | | | - Kristina Ringe
- Institut für Diagnostische und Interventionelle Radiologie, Medizinische Hochschule Hannover
| | - Elke Roeb
- Medizinische Klinik II Pneumologie, Nephrologie und Gastroenterologie, Universitätsklinikum Gießen
| | - Jörn Rüssel
- Medizinische Klinik IV Hämatologie und Onkologie, Universitätsklinikum Halle (Saale)
| | - Barbara Schellhaas
- Medizinische Klinik I Gastroenterologie, Pneumologie und Endokrinologie, Friedrich-Alexander-Universität, Erlangen
| | - Peter Schirmacher
- Allgemeine Pathologie und pathologische Anatomie, Universitätsklinikum Heidelberg
| | | | - Irene Schmid
- Kinderklinik und Kinderpoliklinik im Dr. von Haunerschen Kinderspital, LMU München
| | - Kerstin Schütte
- Klinik für Innere Medizin und Gastroenterologie, Niels-Stensen-Kliniken, Marienhospital Osnabrück
| | - Andreas Schuler
- Medizinische Klinik, Gastroenterologie, Alb-Fils-Kliniken, Geislingen an der Steige
| | - Daniel Seehofer
- Klinik und Poliklinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig
| | - Marianne Sinn
- II. Medizinische Klinik und Poliklinik (Onkologie, Hämatologie, Knochenmarktransplantation mit Abteilung für Pneumologie), Universitätsklinikum Hamburg-Eppendorf
| | - Andreas Stengel
- Innere Medizin VI - Psychosomatische Medizin und Psychotherapie, Eberhard-Karls Universität, Tübingen
| | | | | | | | - Anne Taubert
- Klinische Sozialarbeit, Universitätsklinikum Heidelberg
| | - Jörg Trojan
- Medizinische Klinik 1: Gastroenterologie und Hepatologie, Pneumologie und Allergologie, Endokrinologie und Diabetologie sowie Ernährungsmedizin, Goethe-Universität, Frankfurt
| | | | - Martin Utzig
- Abteilung Zertifizierung, Deutsche Krebsgesellschaft e.V., Berlin
| | - Arndt Vogel
- Institute of Medical Science, University of Toronto
| | - Thomas Vogl
- Institut für Diagnostische und Interventionelle Radiologie, Goethe-Universität, Frankfurt
| | - Frank Wacker
- Institut für Diagnostische und Interventionelle Radiologie, Medizinische Hochschule Hannover
| | | | - Heiner Wedemeyer
- Klinik für Gastroenterologie, Hepatologie, Infektiologie und Endokrinologie, Medizinische Hochschule Hannover
| | - Henning Wege
- Klinik für Allgemeine Innere Medizin, Onkologie/Hämatologie, Gastroenterologie und Infektiologie, Klinikum Esslingen
| | - Gregor Wenzel
- Office des Leitlinienprogrammes Onkologie, Deutsche Krebsgesellschaft e.V., Berlin
| | - Dane Wildner
- Innere Medizin, Krankenhäuser Nürnberger Land GmbH, Standort Lauf
| | - Marcus-Alexander Wörns
- Klinik für Gastroenterologie, Hämatologie und internistische Onkologie und Endokrinologie, Klinikum Dortmund
| | - Peter Galle
- 1. Medizinische Klinik und Poliklinik, Gastroenterologie, Hepatologie, Nephrologie, Rheumatologie, Infektiologie, Johannes Gutenberg-Universität, Mainz
| | - Nisar Malek
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| |
Collapse
|
49
|
Yang M, Yao Y, Wang K, Qi L, Yang B, Khudadad M, Guo Y, Wang Y, Liu Y, Li L, Cao L, Huang Q, Guo Q, Li Q, Yao X, Wang C, Cao W. Clinicopathological characteristics and prognostic significance of HER2 status evaluation in patients with urothelial carcinoma: a retrospective single-center experience in China. Virchows Arch 2025:10.1007/s00428-025-04057-x. [PMID: 40011272 DOI: 10.1007/s00428-025-04057-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/11/2025] [Accepted: 02/16/2025] [Indexed: 02/28/2025]
Abstract
Due to the lack of an in-depth evaluation of urothelial carcinoma (UC) HER2 expression in both primary and recurrent UC, we examined possible clinical and pathological variables associated with altered UC HER2 expression. The HER2 status and clinicopathological features of primary UCs and matched recurrences from 2015 to 2023 (n = 1352) were compared by evolution category (stable or altered). In 1352 UC patients, HER2 protein expression was linked with age, histological grade, and recurrence. HER2 was expressed in 794 UC patients (58.7%) (1 + , 2 + , and 3 +). High-level (HER2 scored 2 + and 3 +) HER2 expression does not correlate with gene amplification. Significant differences in HER2 expression exist between initial and recurrent tumors (p < 0.05), including a shift from positive to negative expression. Recurrent UC patients showed associations between HER2 expression, histological grade (p < 0.05), and time to surgical recurrence (p = 0.008). HER2 expression in high-grade UC correlated positively with PD-L1 expression in tumor cells (TC) (rs = 0.222, p = 0.047) and tumor-associated immune cells (IC) (rs = 0.238, p = 0.032), especially in recurrent patients (rs = 0.464, p = 0.022). HER2 expression was associated with the biological behavior and prognosis of UC patients. Increased HER2 expression in recurrent UC indicates a poorer prognosis. Consequently, retesting for HER2 is essential in cases of recurrent UC. The association between HER2 and PD-L1 expression in high-grade, recurrent UC suggests the potential for utilizing ADC alongside immunotherapy.
Collapse
Affiliation(s)
- Manlin Yang
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Ye Yao
- Department of Biostatistics, University of Michigan, Ann Arbor, USA
| | - Kun Wang
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Lisha Qi
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Bo Yang
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Mahtab Khudadad
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, Dalhousie University, Room 726, 5788 University Avenue, Halifax, NS, B3H 1V8, Canada
| | - Yuhong Guo
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Yalei Wang
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Yanxue Liu
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Lingmei Li
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Lu Cao
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Qiujuan Huang
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Qianru Guo
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Qing Li
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Xin Yao
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Cheng Wang
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, Dalhousie University, Room 726, 5788 University Avenue, Halifax, NS, B3H 1V8, Canada.
| | - Wenfeng Cao
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Tianjin'S Clinical Research Center for Cancer, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China.
| |
Collapse
|
50
|
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
|