1
|
Li X, Lou C, Ren H, Cui L, Chen K. Fundamental knowledge and research regarding the role of immunity in triple-negative breast cancer from 2014-2024: A bibliometric analysis. Hum Vaccin Immunother 2025; 21:2483022. [PMID: 40135819 PMCID: PMC11951696 DOI: 10.1080/21645515.2025.2483022] [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: 09/13/2024] [Revised: 02/27/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Immunity has vital research value and promising applications in triple-negative breast cancer (TNBC). Nevertheless, few bibliometric analyses have systematically investigated this area. This study aimed to comprehensively review the collaboration and impact of countries, institutions, authors, and journals on the role of immunity in TNBC from a bibliometric perspective, evaluate the keyword co-occurrence of the knowledge structure, and identify hot trends and emerging topics. Articles and reviews related to immunity in TNBC were retrieved from the Web of Science core collection using subject search. A bibliometric study was conducted primarily using CiteSpace and VOSviewer. A total of 3,104 articles and reviews were included from January 1, 2014, through December 31, 2024. The number of articles on immunization in TNBC is rising. These publications are mainly from 415 institutions in 82 countries, led by China and the USA. Among these publications, Lajos Pusztai published the most papers, while Peter Schmid was co-cited the most. The most productive journals focused on molecular biology, biological immunology, and clinical medicine. Furthermore, co-citation analysis revealed that tumor microenvironment, biomarkers, and immune checkpoint inhibitors are current and developing research areas. The keywords "immunotherapy" and "nanoparticles" are also likely to be new trends and focal points for future research. This study adopted bibliometric and visualization methods to provide a comprehensive review of the research on immunization in TNBC. This article will help researchers better understand the dynamic evolution of the role of immunity in TNBC and identify areas for future research.
Collapse
Affiliation(s)
- Xudong Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chun Lou
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - He Ren
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lina Cui
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Kexin Chen
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
2
|
Fines C, McCarthy H, Buckley N. The search for a TNBC vaccine: the guardian vaccine. Cancer Biol Ther 2025; 26:2472432. [PMID: 40089851 PMCID: PMC11913391 DOI: 10.1080/15384047.2025.2472432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/17/2025] Open
Abstract
Nearly 20 million people are diagnosed with cancer each year with breast cancer being the most common among women. Triple negative breast cancer (TNBC), defined by its no/low expression of ER and PR and lack of amplification of HER2, makes up 15-20% of all breast cancer cases. While patients overall have a higher response to chemotherapy, this subgroup is associated with the lowest survival rate indicating significant clinical and molecular heterogeneity demanding alternate treatment options. Therefore, new therapies have been explored, with a large focus on utilizing the immune system. A whole host of immunotherapies have been studied including immune checkpoint inhibitors, now standard of care for eligible patients, and possibly the most exciting and promising is that of a TNBC vaccine. While currently there are no approved TNBC vaccines, this review highlights many promising studies and points to an antigen, p53, which we believe is highly relevant for TNBC.
Collapse
Affiliation(s)
- Cory Fines
- School of Pharmacy, Queen’s University Belfast, Belfast, UK
| | - Helen McCarthy
- School of Pharmacy, Queen’s University Belfast, Belfast, UK
| | - Niamh Buckley
- School of Pharmacy, Queen’s University Belfast, Belfast, UK
| |
Collapse
|
3
|
Barış Moğul C, Duran MB, Caner V, Türk NŞ, Tuncay ÖL. The PD-L1 Promoter Methylation Predicts Gene And Protein Expression Levels in Urothelial Carcinoma. Gene 2025; 959:149503. [PMID: 40228759 DOI: 10.1016/j.gene.2025.149503] [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: 11/26/2024] [Revised: 04/07/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
We aimed to clarify the role of PD-L1 promoter methylation in bladder cancer by analyzing PD-L1 methylation and mRNA expression in FFPE samples, along with PD-L1 mRNA and protein levels in urine samples from bladder urothelial carcinoma patients. We analyzed PD-L1 promoter methylation in 43 bladder urothelial carcinoma tissue samples and 41 non-malignant bladder tissues using methylation-sensitive high-resolution melting analysis to assess two CpG islands (cg15837913, cg19724470). PD-L1 mRNA expression in tissues and urine samples, along with PD-L1 protein levels in urine, were evaluated. The bladder urothelial carcinoma group showed significantly higher methylation rates for cg19724470 and cg15837913 compared to controls (P = 0.016, P = 0.049 respectively). In the patient group, tissue PD-L1 mRNA expression was 15.22 times higher and urinary PD-L1 mRNA expression 6.56 times higher in the cg19724470 unmethylated group compared to the methylated group. Urinary PD-L1 protein concentration was twice as high in the cg19724470 unmethylated group compared to the methylated group. In the patients, tissue PD-L1 mRNA expression was 4.58 times higher and urinary PD-L1 mRNA expression 2.58 times higher in the cg15837913 unmethylated group compared to the methylated group. Moreover, the urinary PD-L1 protein concentration was 1.7 times higher in the cg15837913 unmethylated group than in the methylated group (P = 0.036). A positive correlation was observed between tissue PD-L1 mRNA and both urine PD-L1 mRNA and protein levels and between urine PD-L1 mRNA and protein levels. This study suggests that PD-L1 methylation may be a key epigenetic regulator influencing PD-L1 expression and disease pathogenesis in bladder urothelial carcinoma.
Collapse
Affiliation(s)
- Cansu Barış Moğul
- Department of Medical Biology, School of Medicine, Pamukkale University, Denizli, Turkey.
| | - Mesut Berkan Duran
- Department of Urology, School of Medicine, Pamukkale University, Denizli, Turkey.
| | - Vildan Caner
- Department of Medical Genetics, School of Medicine, Pamukkale University, Denizli, Turkey; Sapiens Genetics Diagnostic Center, İstanbul, Turkey.
| | - Nilay Şen Türk
- Department of Medical Pathology, School of Medicine, Pamukkale University, Denizli, Turkey.
| | - Ömer Levent Tuncay
- Department of Urology, School of Medicine, Pamukkale University, Denizli, Turkey
| |
Collapse
|
4
|
Zhang Z, Zhao Q, Xu Q, Deng Q, Hua A, Wang X, Yang X, Li Z. A mitochondria-interfering nanocomplex cooperates with photodynamic therapy to boost antitumor immunity. Biomaterials 2025; 317:123094. [PMID: 39799701 DOI: 10.1016/j.biomaterials.2025.123094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
Immunotherapeutics against triple-negative breast cancer (TNBC) hold great promise. In this work, we provide a combination therapy for simultaneous increasing tumor immunogenicity and down-regulating programmed cell death ligand 1 (PD-L1) to boost antitumor immunity in TNBC. We prepare bis (diethyldithiocarbamate)-copper/indocyanine green nanoparticles (CuET/ICG NPs) simply in aqueous with one-pot method. CuET/ICG NPs interfere mitochondria, reduce oxygen consumption, and alleviate tumor hypoxia to potentiate photodynamic therapy (PDT) for amplifying immunogenic cell death (ICD). Meanwhile, mitochondria dysfunction leads to energy stress and activates AMPK pathway. As a result, CuET/ICG NPs downregulates membrane PD-L1 (mPD-L1) on both 4T1 cancer cells and cancer stem cells (CSCs) through AMP-activated protein kinase (AMPK)-mediated pathway in hypoxia. Cooperatively, the combinational therapy activates antitumor immunity and triggers long lasting immune memory response to resist tumor re-challenge. Our study represents an attempt that conquers tumor immunosuppressive microenvironment with simple biomedical materials and multimodality treatments.
Collapse
Affiliation(s)
- Zhijie Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Qingfu Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Qingqing Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Qingyuan Deng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Ao Hua
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Xing Wang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, PR China.
| | - Zifu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, PR China.
| |
Collapse
|
5
|
Kim ES. Molecular targets and therapies associated with poor prognosis of triple‑negative breast cancer (Review). Int J Oncol 2025; 66:52. [PMID: 40444482 PMCID: PMC12118953 DOI: 10.3892/ijo.2025.5758] [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: 03/15/2025] [Accepted: 05/07/2025] [Indexed: 06/02/2025] Open
Abstract
Triple‑negative breast cancer (TNBC) is a highly aggressive and heterogeneous subtype of BC characterized by the absence of estrogen, progesterone and human EGFR2 receptors. This lack of receptors renders it unresponsive to standard targeted therapies. Despite advances made in understanding the molecular landscape of TNBC, its poor prognosis and high recurrence rates underscore the urgent need for innovative therapeutic approaches. This review explores the effects of key prognostic markers, such as Ki‑67, programmed cell death ligand 1, BRCA1/2 mutations, E‑cadherin loss and EGFR alterations. It also examines critical pathways, including the PI3K/AKT/mTOR and mutant p53 pathways, which are prerequisites for TNBC progression and therapy resistance, and discusses the therapeutic potential of directly targeting these key molecules and their associated signaling pathways. In addition, recent advances in targeted therapies were highlighted, such as immune checkpoint inhibitors, and the statuses of emerging strategies were presented, such as chimeric antigen receptor‑T cell therapy and small inhibitory RNA‑based treatments. Given the molecular heterogeneity of TNBC, the importance of precision medicine was also discussed and it was emphasized that this approach is becoming an increasingly critical aspect of personalized treatment strategies. Resistance to existing therapies presents a major challenge to the effective treatment of TNBC, and thus, the development of future therapeutic strategies requires technical innovations. By integrating these insights, this review aims to provide a comprehensive overview of current and future means of improving TNBC outcomes.
Collapse
Affiliation(s)
- Eun-Sook Kim
- College of Pharmacy, Duksung Women's University, Seoul 01369, Republic of Korea
| |
Collapse
|
6
|
Fu Y, Yang Q, Xu N, Zhang X. MiRNA affects the advancement of breast cancer by modulating the immune system's response. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167759. [PMID: 40037267 DOI: 10.1016/j.bbadis.2025.167759] [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: 10/29/2024] [Revised: 02/05/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Breast cancer (BC), which is the most common tumor in women, has greatly endangered women's lives and health. Currently, patients with BC receive comprehensive treatments, including surgery, chemotherapy, radiotherapy, endocrine therapy, and targeted therapy. According to the latest research, the development of BC is closely related to the inflammatory immune response, and the immunogenicity of BC has steadily been recognized. As such, immunotherapy is one of the promising and anticipated forms of treatment for BC. The potential values of miRNA in the diagnosis and prognosis of BC have been established, and aberrant expression of associated miRNA can either facilitate or inhibit progression of BC. In the tumor immune microenvironment (TME), miRNAs are considered to be an essential molecular mechanism by which tumor cells interact with immunocytes and immunologic factors. Aberrant expression of miRNAs results in reprogramming of tumor cells actively, which may suppress the generation and activation of immunocytes and immunologic factors, avoid tumor cells apoptosis, and ultimately result in uncontrolled proliferation and deterioration. Therefore, through activating and regulating the immunocytes related to tumors and associated immunologic factors, miRNA can contribute to the advancement of BC. In this review, we assessed the function of miRNA and associated immune system components in regulating the advancement of BC, as well as the potential and viability of using miRNA in immunotherapy for BC.
Collapse
Affiliation(s)
- Yeqin Fu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China
| | - Qiuhui Yang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 310006, China
| | - Ning Xu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Xiping Zhang
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
7
|
Jain M, Jadhav IM, Dangat SV, Singuru SR, Sethi G, Yuba E, Gupta RK. Overcoming the novel glycan-lectin checkpoints in tumor microenvironments for the success of the cross-presentation-based immunotherapy. Biomater Sci 2025. [PMID: 40421610 DOI: 10.1039/d4bm01732c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
In pursuit of meeting the ever-rising demand for cancer therapies, cross-presentation-based glyconanovaccines (GNVs) targeting C-type lectin receptors (CLRs) on DCs have shown significant potential as cutting-edge cancer immunotherapy. GNVs are an attractive approach to induce anti-cancer cytotoxic T lymphocyte responses. Despite immune checkpoints (ICs) being well established and an obstacle to the success of GNVs, glycan-lectin circuits are emerging as unique checkpoints due to their immunomodulatory functions. Given the role of aberrant tumor glycosylation in promoting immune evasion, mitigating these effects is crucial for the efficacy of GNVs. Lectins, such as siglecs and galectins, are detrimental to the tumor immune landscape as they promote an immunosuppressive TME. From this perspective, this review aims to explore glycan-lectin ICs and their influence on the efficacy of GNVs. We aim to discuss various ICs in the TME followed by drawbacks of immune checkpoint inhibitors (ICIs). We will also emphasize the altered glycosylation profile of tumors, addressing their immunosuppressive nature along with ways in which CLRs, siglecs, and galectins contribute to immune evasion and cancer progression. Considering the resistance towards ICIs, current and prospective approaches for targeting glycan-lectin circuits and future prospects of these endeavors in harnessing the full potential of GNVs will also be highlighted.
Collapse
Affiliation(s)
- Mannat Jain
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Isha M Jadhav
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Suyash Vinayak Dangat
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Srinivasa Rao Singuru
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore.
| | - Eiji Yuba
- Department of Chemistry & Bioengineering, Graduate School of Engineering, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka-city, Osaka 558-8585, Japan.
| | - Rajesh Kumar Gupta
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| |
Collapse
|
8
|
Tiberi E, Parisi A, Pistelli M, Savini A, Galassi F, Reschini C, Quintavalle D, Napoleoni R, Ferrari C, Berardi R. Immunotherapy in Triple-Negative Breast Cancer. Oncol Ther 2025:10.1007/s40487-025-00346-2. [PMID: 40418298 DOI: 10.1007/s40487-025-00346-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 04/29/2025] [Indexed: 05/27/2025] Open
Abstract
Currently, immunotherapy has led to a paradigmatic shift in the treatment of many cancer types, including triple-negative breast cancer. Immunotherapy increases the efficacy of the immune system in treating cancer, with a durable effect due to immunologic memory. The PD-1 inhibitor, pembrolizumab, combined with neoadjuvant chemotherapy, improved event-free survival and is a new standard of care for patients with high-risk, early stage triple-negative breast cancer (TNBC), regardless of tumor PD-L1 expression. For metastatic TNBC, pembrolizumab combined with chemotherapy is a new standard of care for first-line therapy for PD-L1+ metastatic TNBC, and it improves overall survival. The PD-L1 inhibitor, atezolizumab, combined with nab-paclitaxel, is also approved for first-line treatment of metastatic PD-L1+ TNBC. The aim of this review is to examine the existing evidence and ongoing studies on immunotherapy in patients with early stage and metastatic triple-negative breast cancer (TNBC), including new combination strategies with several drugs, such as chemotherapy, targeted therapy, or radiation and to discuss immune checkpoint inhibitor (ICI) applications and the possibility of emerging strategies in different TNBC stages.
Collapse
Affiliation(s)
- Elisa Tiberi
- Clinica Oncologica, Università Politecnica delle Marche, AOU delle Marche, Ancona, Italy.
| | - Alessandro Parisi
- Clinica Oncologica, Università Politecnica delle Marche, AOU delle Marche, Ancona, Italy.
| | - Mirco Pistelli
- Clinica Oncologica, Università Politecnica delle Marche, AOU delle Marche, Ancona, Italy
| | - Agnese Savini
- Clinica Oncologica, Università Politecnica delle Marche, AOU delle Marche, Ancona, Italy
| | | | | | | | | | | | - Rossana Berardi
- Clinica Oncologica, Università Politecnica delle Marche, AOU delle Marche, Ancona, Italy
| |
Collapse
|
9
|
Zhong X, Han J, Li H, Shen X, Yu B, Chen T, Li H, Li J, Pang J, Qian L, Wu W, Tong X, Ding B. Glycosylated protein-related microenvironmental features in breast cancer are associated with patient prognosis. Mamm Genome 2025:10.1007/s00335-025-10137-9. [PMID: 40411577 DOI: 10.1007/s00335-025-10137-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Accepted: 05/10/2025] [Indexed: 05/26/2025]
Abstract
The tumor microenvironment (TME) and aberrant glycosylation have been suggested to play key roles in cancer. This study integrated differentially expressed genes (DEGs) and weighted gene coexpression network analysis (WGCNA) to identify tumor microenvironment-related genes and construct a TME-risk prognostic signature (TMERS) through LASSO Cox regression. After batch effect removal, 44 TME-prognosis-related genes (TMEPGs) were identified and classified into three molecular subtypes via K-means clustering. The finalized 22-gene TMERS model demonstrated robust prognostic predictive capacity in GEO datasets. The results revealed distinct immune profiles and prognostic stratifications among genetic subtypes and risk groups, confirming that the TMERS is an independent prognostic indicator for breast cancer (BRCA). Glycosyltransferase genes (GTs) have potential therapeutic relevance through immune regulation, with TMEPG member killer cell lectin like receptor B1 (KLRB1) significantly correlated with BRCA prognosis. Cellular experiments demonstrated that KLRB1 overexpression suppressed BRCA cell proliferation and migration. This work establishes a novel prognostic model for BRCA while highlighting KLRB1 as a potential biomarker, providing new insights into TME-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoxiao Zhong
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
| | - Jiaxuan Han
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Huan Li
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Xiangyu Shen
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Bowen Yu
- Department of Gastrointestinal Surgery, Third XiangYa Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Ting Chen
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Haobing Li
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, China
| | - Jun Li
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Jin Pang
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Liyuan Qian
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Wei Wu
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Xiaoliang Tong
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, 410000, China.
| | - Boni Ding
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
| |
Collapse
|
10
|
He L, He J, Jiang T, Gong R, Wan X, Duan M, Chen Z, Cheng Y. Inhibition of UCH-L1 enhances immunotherapy efficacy in triple-negative breast cancer by stabilizing PD-L1. Eur J Pharmacol 2025; 1000:177743. [PMID: 40389130 DOI: 10.1016/j.ejphar.2025.177743] [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: 01/28/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Recent research indicates that programmed death 1 (PD-1) and programmed death-ligand 1 (PD-L1) inhibitors show promise in treating triple-negative breast cancer (TNBC), but their efficacy is lower than anticipated, especially when used alone. Therefore, enhancing the anti-tumor immune response strategy for TNBC is crucial. Ubiquitin carboxy-terminal hydrolase L1 (UCH-L1), involved in tumor cell regulation and a potential therapeutic target, has an undefined role in TNBC immunotherapy. In this study, we explored the inverse correlation between UCH-L1 and PD-L1 in TNBC patient tissues. Through in vitro experiments, we found that UCH-L1 negatively regulates PD-L1 by stabilizing the E3 ubiquitin ligase ariadne-1 homolog (ARIH1), which promotes PD-L1 ubiquitination and degradation. Further analysis in Balb/c mice xenograft tumors showed that UCH-L1 correlates with GZMB+/CD8+ T cell infiltration in TNBC, suggesting potential synergistic effects when combining UCH-L1 inhibitors with PD-L1 antibodies. Overall, in TNBC, UCH-L1 stabilizes ARIH1, leading to low PD-L1 expression, which may explain the limited effectiveness of immunotherapy in TNBC patients. Our mouse experiments showed improved therapeutic effects when combining UCH-L1 inhibitors with PD-L1 antibodies. These findings offer a new avenue for immunotherapy in TNBC patients.
Collapse
Affiliation(s)
- Linhao He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Jiaying He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ting Jiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Rong Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Xiaoya Wan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Mingwu Duan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Zonglin Chen
- Clinical Research Center for Breast Disease in Hunan Province, Changsha, 410011, China
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China; Clinical Research Center for Breast Disease in Hunan Province, Changsha, 410011, China; NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, 410008, China; Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Changsha, 410011, China.
| |
Collapse
|
11
|
Wilkerson AD, Juric I, Singh S, Rayman PA, Pavicic PG, Powers J, Parthasarathy PB, Al-Hilli Z, Ko JS, Chan T, Alban T, Montero AJ, Diaz-Montero CM. Responses to checkpoint inhibition in metastatic triple negative breast cancer driven by divergent myeloid phenotypes. COMMUNICATIONS MEDICINE 2025; 5:180. [PMID: 40382442 PMCID: PMC12085700 DOI: 10.1038/s43856-025-00860-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 04/10/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Given the scarcity of effective therapeutic targets, metastatic triple negative breast cancer (mTNBC) has shorter survival times compared to other advanced breast cancer subtypes. Although chemo-immunotherapy with immune checkpoint inhibitors (ICIs) in PD-L1+ mTNBC has shown promise, survival benefit remains modest. Therefore, it is crucial to gain improved insight into the mechanisms underlying response and resistance to checkpoint inhibition in mTNBC. METHODS We employed single cell RNA sequencing (scRNAseq), single cell secretomics, and flow cytometry to identify transcriptomic and proteomic peripheral immune cell signatures associated with response and non-response to anti-PD-1/PD-L1 therapy and chemotherapy in mTNBC. RESULTS Transcriptomic analysis reveal divergent transcriptional programming of CD33+ myeloid cells between responders and non-responders, even in pretreatment PBMC samples. This divergence, in responders, is characterized by an immune-promoting CD33+ cell phenotype involving IL1b signaling compared to non-responders, where an immunosuppressive phenotype marked by IL1b inhibition is observed. These baseline differences become more pronounced during the course of chemo-immunotherapy. Differences in CD33+ cell phenotype result in functional differences in lymphocyte activities between responders and non-responders. Depletion of CD33+ cells in pre-treatment samples from non-responders, restores T cell effector function. CONCLUSION Our findings highlight CD33+ cell phenotype as a key determinant of response to chemo-immunotherapy, which can be assessed from peripheral blood. This offers a valuable tool in the context of metastatic TNBC, in which tissue sampling is often challenging.
Collapse
Affiliation(s)
- Avia D Wilkerson
- Cleveland Clinic Foundation, Center for Immunotherapy & Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH, USA
- Cleveland Clinic Foundation, Department of General Surgery, Digestive Disease & Surgery Institute, Cleveland, OH, USA
| | - Ivan Juric
- Cleveland Clinic Foundation, Center for Immunotherapy & Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH, USA
| | - Salendra Singh
- Cleveland Clinic Foundation, Center for Immunotherapy & Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH, USA
| | - Patricia A Rayman
- Cleveland Clinic Foundation, Center for Immunotherapy & Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH, USA
| | - Paul G Pavicic
- Cleveland Clinic Foundation, Center for Immunotherapy & Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH, USA
| | - Jennifer Powers
- Cleveland Clinic Foundation, Center for Immunotherapy & Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH, USA
| | - Prerana Bangalore Parthasarathy
- Cleveland Clinic Foundation, Center for Immunotherapy & Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH, USA
| | - Zahraa Al-Hilli
- Cleveland Clinic Foundation, Department of General Surgery, Digestive Disease & Surgery Institute, Cleveland, OH, USA
| | - Jennifer S Ko
- Cleveland Clinic Foundation, Pathology and Laboratory Medicine Institute, Cleveland, OH, USA
| | - Timothy Chan
- Cleveland Clinic Foundation, Center for Immunotherapy & Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH, USA
| | - Tyler Alban
- Cleveland Clinic Foundation, Center for Immunotherapy & Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH, USA.
| | - Alberto J Montero
- University Hospitals/Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
| | - C Marcela Diaz-Montero
- Cleveland Clinic Foundation, Center for Immunotherapy & Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH, USA.
| |
Collapse
|
12
|
Yao P, Ju H, Song A, Wang Y, Xin G, Wang G, Ma J, Guo M. Ruxolitinib suppresses tumor growth in PTEN-deficient glioblastoma by inhibiting the STAT3-PDL1 axis-mediated the M2 polarization of macrophages. Int Immunopharmacol 2025; 155:114629. [PMID: 40239334 DOI: 10.1016/j.intimp.2025.114629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/29/2025] [Accepted: 04/05/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Glioblastoma (GBM) is the most malignant form of brain tumor, and GBM patients with poorer prognosis and highly immunosuppressive tumor microenvironment (TME) often exhibit PTEN deficiency in their tumor tissues. Therefore, new therapeutic strategies targeting immunosuppressive TME maybe useful in PTEN-deficient GBM. METHODS Bioinformatics was used to assess gene expression, survival time and immunoinfiltration in PTEN-deficient GBM. CRISPR-Cas9 was used to construct gene knockout cell lines. C57BL/6 mouse orthotopic GBM models were used to conduct survival analysis and evaluate treatment effect of Ruxolitinib. Flow cytometry, immunohistochemistry, immunofluorescence and quantitative real-time PCR (qRT-PCR) to detect the polarization of macrophages. Immunoblotting, immunohistochemistry, qRT-PCR, enzyme linked immunosorbent assay, and dual-luciferase reporter assay were used to conduct mechanism research. RESULTS We identified that the elevated levels of phosphorylated STAT3 (p-STAT3) in PTEN-deficient GBM facilitate PDL1 transcription, which subsequently drives M2 polarization of macrophages. Furthermore, PTEN deficiency, along with high expression levels of STAT3 and PDL1, are associated with a shorter survival time in GBM patients. Notably, in orthotopic mouse models of GBM with PTEN deficiency, Ruxolitinib therapy reduces the levels of p-STAT3 and PDL1, inhibits the infiltration of M2 macrophages, and suppresses tumor growth. CONCLUSIONS The STAT3-PDL1 axis plays a crucial role in the M2 polarization of macrophages in PTEN-deficient GBM. The blockade of the STAT3-PDL1 axis by Ruxolitinib regulates the anti-tumor immune response and curtails tumor progression in PTEN-deficient GBM, highlighting its significant clinical implications.
Collapse
Affiliation(s)
- Penglei Yao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huanyu Ju
- Department of Immunology, Harbin Medical University, Harbin, China
| | - Aohua Song
- Department of Immunology, Harbin Medical University, Harbin, China
| | - Yue Wang
- Department of Immunology, Harbin Medical University, Harbin, China
| | - Guoshun Xin
- Department of Immunology, Harbin Medical University, Harbin, China
| | - Guangzhi Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jian Ma
- Department of Immunology, Harbin Medical University, Harbin, China; Department of Hepatopancreatobiliary, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Mian Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
13
|
Xu T, Zhang H, Yang BB, Qadir J, Yuan H, Ye T. Tumor-infiltrating immune cells state-implications for various breast cancer subtypes. Front Immunol 2025; 16:1550003. [PMID: 40438111 PMCID: PMC12116345 DOI: 10.3389/fimmu.2025.1550003] [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: 12/22/2024] [Accepted: 04/24/2025] [Indexed: 06/01/2025] Open
Abstract
Breast cancer presents a variety of subtypes due to its cellular and molecular heterogeneity. The capacity of cancer cells to proliferate, invade, and metastasize depends not only on their intrinsic characters but also on their dynamic interaction with the host tumor microenvironment (TME), which includes immune cells. Meanwhile, the infiltration of immune cells in the TME severely affects the occurrence, development, treatment, and prognosis of breast cancer. Therefore, this review aims to explore the immune invasive tumor microenvironment in different intrinsic subtypes of breast cancer. Additionally, it highlights the mechanistic influence of the infiltrating immune cells on stage-wise dynamics of breast tumorigenesis. Moreover, the present review also attempts to discern the regulatory relationship between tumor infiltrating immune cells and immune microenvironment in different molecular subtypes of breast cancer, thus, spotlighting its clinical significance.
Collapse
Affiliation(s)
- Tianshuang Xu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Hongjun Zhang
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Burton B. Yang
- Sunnybrook Research Institute and the Department of Laboratory Medicine and Pathobiology at the University of Toronto, Toronto, ON, Canada
| | - Javeria Qadir
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Hui Yuan
- School of Stomatology and School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Ting Ye
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
14
|
Wiertsema P, Tan YH, Haanen JBAG, Seijkens TTP, Jedema I. Advances in TIL therapy: Expanding the horizons beyond melanoma. MED 2025:100702. [PMID: 40381620 DOI: 10.1016/j.medj.2025.100702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/08/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Abstract
Tumor-infiltrating lymphocyte (TIL) therapy represents a breakthrough in solid tumor treatment, addressing unmet needs for patients with limited options. While its efficacy is established in advanced melanoma, TIL therapy shows early promise in non-small cell lung cancer, breast cancer, gynecological cancers, and head and neck cancers. However, challenges such as reduced T cell infiltration, lower tumor mutational burden (TMB), immunosuppressive tumor microenvironments (TME), and toxicity associated with the TIL therapy regimen hinder its broader application in these patient groups, compared with melanoma. To address these challenges, new approaches focus on the selection of tumor-reactive TIL, optimization of TIL expansion, combination of immune checkpoint inhibitors with TIL therapy to counteract immunosuppressive microenvironments, and genetic modification of TIL to enhance persistence and functionality. Larger clinical trials are essential to validate these innovations and standardize protocols. With continued advancements, TIL therapy has the potential to redefine the treatment landscape for advanced solid cancers.
Collapse
Affiliation(s)
- Pauline Wiertsema
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ya Hwee Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore; Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - John B A G Haanen
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands; Melanoma Clinic, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Tom T P Seijkens
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Inge Jedema
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
15
|
Stopeck AT, Abu-Khalaf M, Borges V, Chmielowski B, Rao R, Xie B, Dudek AZ, Mina L, O'Shaughnessy J, Chisamore M, Mattson P, Gargano M, Cox J, Osterwalder B, Drees J, Harrison B, Chan ASH, Qiu X, Ottoson N, Bose N, Uhlik M, Graff J, Iglesias J. Phase 2 trial of imprime and pembrolizumab immunotherapy in metastatic triple negative breast cancer patients who have progressed beyond first line chemotherapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf079. [PMID: 40338159 DOI: 10.1093/jimmun/vkaf079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 03/12/2025] [Indexed: 05/09/2025]
Abstract
The Phase 2 IMPRIME 1 study evaluated the combination of the pathogen-associated molecular pattern (PAMP) Imprime with the immune checkpoint inhibitor (ICI) pembrolizumab as second or later line of treatment (2 L+) for patients with metastatic triple-negative breast cancer (mTNBC). Eligible patients with mTNBC received weekly Imprime (4 mg/kg) intravenously in combination with pembrolizumab (200 mg every 3 weeks). Primary endpoints were overall response rate (ORR) and safety. Secondary endpoints included disease control rate (DCR), duration of response (DoR), progression free survival (PFS), and overall survival (OS). Exploratory endpoints included correlations between immune cell activation markers in tumor tissues and blood and response to therapy. Of the 42 evaluable patients, six had a response (one complete, five partial), with an ORR of 14.3% by RECIST v1.1. Median PFS was 2.7 months, median OS was 16.4 months, and DCR was 54.8%, with responders achieving a median DoR of 15.2 months. Therapy was generally well tolerated and resulted in an increase of immune activation markers, with higher levels of activation in peripheral blood associated with response and improved survival. The combination of Imprime and pembrolizumab was safe and demonstrated immune activation in tumor tissues and peripheral blood in patients with TNBC. Improved response rates were observed compared to historical studies of ICI monotherapy in similar patient populations. Study number (ClinicalTrials.gov trial registration): NCT02981303.
Collapse
Affiliation(s)
- Alison T Stopeck
- Department of Hematology and Oncology, Stony Brook University, Stony Brook, NY, United States
| | | | - Virginia Borges
- Medicine-Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Bartosz Chmielowski
- Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA, United States
| | - Ruta Rao
- Rush University Medical Center, Chicago, IL, United States
| | - Bin Xie
- Hematology, Hematology Oncology, Medical Oncology, Swedish Cancer Institute, Issaquah, WA, United States
| | | | - Lida Mina
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
- Banner MD Anderson Cancer Center, Phoenix, AZ, United States
| | - Joyce O'Shaughnessy
- Medical Oncology, Internal Medicine, Baylor University, Medical Center, Texas Oncology, US Oncology, Dallas, TX, United States
| | | | | | | | - Joanna Cox
- Zentalis Pharmaceutical, San Diego, CA, United States
| | | | | | | | | | | | | | | | | | | | - Jose Iglesias
- APEX Oncology Consulting, Inc., Oakville, ON, Canada
| |
Collapse
|
16
|
Elder AM, Fairchild HR, Kines KT, Cozzens LM, Becks AR, Slansky JE, Anderson SM, Lyons TR. Semaphorin7A and PD-L1 cooperatively drive immunosuppression during mammary involution and breast cancer. Cell Rep 2025; 44:115676. [PMID: 40333186 DOI: 10.1016/j.celrep.2025.115676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/06/2025] [Accepted: 04/16/2025] [Indexed: 05/09/2025] Open
Abstract
Postpartum mammary gland remodeling after a pregnancy/lactation cycle is characterized by mechanisms of cell death and inflammation. Here, we show that SEMA7A promotes PD-L1 expression in immune cells of the mammary tissue during involution. These same phenotypes are mimicked in the microenvironment of SEMA7A-expressing tumors, which partially respond to αPD-1/αPD-L1 treatments in vivo. However, cells that remain after treatment are enriched for SEMA7A expression. Therefore, we tested a monoclonal antibody that directly targets SEMA7A-expressing tumors, in part, by reducing SEMA7A-mediated upregulation of PD-L1. In vivo, the SEMA7A monoclonal antibody reduces tumor growth and/or promotes complete regression of mouse mammary tumors, reduces some immunosuppressive phenotypes in the tumor microenvironment, and restores cytotoxic T cells, suggesting that SEMA7A may be a candidate for immune-based therapy for breast cancer patients.
Collapse
Affiliation(s)
- Alan M Elder
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Cancer Biology Graduate Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Heather R Fairchild
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kelsey T Kines
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Cancer Biology Graduate Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lauren M Cozzens
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Cancer Biology Graduate Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alexandria R Becks
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Cancer Biology Graduate Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jill E Slansky
- University of Colorado Cancer Center, Aurora, CO, USA; Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Traci R Lyons
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; University of Colorado Cancer Center, Aurora, CO, USA; Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
17
|
Dai YW, Wu ZX, Cheng Y, Wu HD, Chen JW, Lv LX, Wang ZQ, Li HF, Yan CZ, Bao JX, Liu CH, Dai XX. Formosanin C inhibits triple-negative breast cancer progression by suppressing the phosphorylation of STAT3 and the polarization of M2 macrophages. Breast Cancer Res Treat 2025; 211:71-89. [PMID: 39953272 DOI: 10.1007/s10549-025-07623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 01/20/2025] [Indexed: 02/17/2025]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC), is a highly aggressive tumor. Formosanin C (FC) is a diosgenin with immunomodulatory and antitumor properties, the precise mechanism through which it is against TNBC remains uncertain. OBJECTIVE Clarifying the mechanism of FC against TNBC. MATERIALS AND METHODS The impact of FC on two TNBC cell lines for 24 h was investigated through various techniques including the CCK8 assay, flow cytometry, transwell assay, scratch tests, immunoblot assay, and immunofluorescence. To elucidate the mechanism behind the anti-TNBC effect of FC, MDA-MB-231 cells were subjected to STAT3 overexpression. Moreover, the in vivo efficacy of FC was examined using a xenograft nude mice (BALB/C). Mice were divided into the control group (equal amount of PBS), the napabucasin group (5 mg/kg) and the FC groups (1 mg/kg, 2 mg/kg). The study duration was 30 days. RESULTS FC exhibited inhibitory effects against MDA-MB-231 and Hs578T cells. FC can decrease the migratory capacity of TNBC cells by inhibiting epithelial-mesenchymal transition (EMT). Meanwhile, we demonstrated that the inhibition of phosphorylation of STAT3 (Y705) is the crucial mechanism of FC against TNBC. Moreover, FC also hindered the polarization of macrophage M2. DISCUSSION AND CONCLUSION This study is the first to show that FC restrains the EMT of TNBC cells by obstructing the STAT3 pathway and hinders the M2 polarization of macrophages and immune evasion. Therefore, FC holds the possibility of being utilized as a therapeutic remedy for TNBC.
Collapse
Affiliation(s)
- Yin-Wei Dai
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhi-Xuan Wu
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yao Cheng
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hao-Dong Wu
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jia-Wei Chen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lin-Xi Lv
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zi-Qiong Wang
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hong-Feng Li
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cong-Zhi Yan
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing-Xia Bao
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cong-Hui Liu
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuan-Xuan Dai
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
18
|
Wang X, Wang L, Liu Y. Current Status of Immune Checkpoint Inhibitors and Treatment Responsive Biomarkers for Triple-Negative Breast Cancer. Thorac Cancer 2025; 16:e70072. [PMID: 40324951 PMCID: PMC12052518 DOI: 10.1111/1759-7714.70072] [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: 12/16/2024] [Revised: 03/24/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025] Open
Abstract
Triple-negative breast cancer (TNBC), accounting for about 10%-20% of all breast cancer cases, is characterized by its aggressive nature, high recurrence rates, and poor prognosis. Unlike other breast cancer subtypes, TNBC lacks hormone receptors and specific molecular targets, limiting therapeutic options. In recent years, immune checkpoint inhibitors (ICIs) have shown promise in treating TNBC by targeting immune evasion mechanisms. Despite these advancements, several issues remain unresolved, including low response rates in programmed cell death ligand 1 (PD-L1) negative TNBC subtypes and the challenge of predicting which patients will benefit from ICIs. Consequently, there is growing interest in identifying reliable biomarkers beyond PD-L1 expression. This review synthesizes recent studies to provide a comprehensive perspective on ICI therapy in TNBC, clarifying the status of single-agent ICI therapies and combination strategies, emphasizing the need for further research into biomarkers. These insights provide clues for more personalized and effective treatment approaches, ultimately aiming to improve clinical outcomes for patients with TNBC.
Collapse
Affiliation(s)
- Xinran Wang
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Lingxia Wang
- Value & Implementation, Global Medical & Scientific AffairsMSD ChinaShanghaiChina
| | - Yueping Liu
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| |
Collapse
|
19
|
Coleman C, Selvakumar T, Thurlapati A, Graf K, Pavuluri S, Mehrotra S, Sahin O, Sivapiragasam A. Harnessing Tumor-Infiltrating Lymphocytes in Triple-Negative Breast Cancer: Opportunities and Barriers to Clinical Integration. Int J Mol Sci 2025; 26:4292. [PMID: 40362529 PMCID: PMC12072607 DOI: 10.3390/ijms26094292] [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: 03/21/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Triple-negative breast cancer (TNBC) continues to present a therapeutic challenge due to the fact that by definition, these cancer cells lack the expression of targetable receptors. Current treatment options include cytotoxic chemotherapy, antibody-drug conjugates (ADC), and the PD-1 checkpoint inhibitor, pembrolizumab. Due to high rates of recurrence, current guidelines for early-stage TNBC recommend either multi-agent chemotherapy or chemo-immunotherapy in all patients other than those with node-negative tumors < 0.5 cm. This approach can lead to significant long-term effects for TNBC survivors, driving a growing interest in de-escalating therapy where appropriate. Tumor infiltrating lymphocytes (TILs) represent a promising prognostic and predictive biomarker for TNBC. These diverse immune cells are present in the tumor microenvironment and within the tumor itself, and multiple retrospective studies have demonstrated that a higher number of TILs in early-stage TNBC portends a favorable prognosis. Research has also explored the potential of TIL scores to predict the response to immunotherapy. However, several barriers to the widespread use of TILs in clinical practice remain, including logistical and technical challenges with the scoring of TILs and lack of prospective trials to validate the trends seen in retrospective studies. This review will present the current understanding of the role of TILs in TNBC and discuss the future directions of TIL research.
Collapse
Affiliation(s)
- Cara Coleman
- Department of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA (T.S.); (S.P.)
| | - Tharakeswari Selvakumar
- Department of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA (T.S.); (S.P.)
| | - Aswani Thurlapati
- Department of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA (T.S.); (S.P.)
| | - Kevin Graf
- Department of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA (T.S.); (S.P.)
| | - Sushma Pavuluri
- Department of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA (T.S.); (S.P.)
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ozgur Sahin
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Abirami Sivapiragasam
- Department of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA (T.S.); (S.P.)
| |
Collapse
|
20
|
Zhang Y, Yang H, Jiang Y, Jiang Y, Mao R. Angiogenesis and immune microenvironment in triple-negative breast cancer: Targeted therapy. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167880. [PMID: 40316057 DOI: 10.1016/j.bbadis.2025.167880] [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: 03/15/2025] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype that typically lacks effective targeted therapies, leading to limited treatment options. Chemotherapy remains the primary treatment modality; however, in recent years, new immunotherapy approaches, such as immune checkpoint inhibitors, have shown positive results in some patients. Although the development of TNBC is closely associated with BRCA gene mutations, the tumor immune microenvironment (TIME) plays a crucial role in tumor progression and immune escape. Tumor angiogenesis, the accumulation of immunosuppressive cells, and alterations in immune molecules collectively shape an environment unfavorable for anti-tumor immune responses. Tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) promote immune escape by secreting immunosuppressive factors. Therefore, combination strategies of anti-angiogenic and immune checkpoint inhibitory therapies have shown synergistic effects in clinical trials, while new targeted therapies such as TGF-β inhibitors and IL-1β inhibitors offer new options for TNBC treatment. With the development of personalized medicine, combining immunotherapy and targeted therapies brings new hope for TNBC patients.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pathophysiology, School of Medicine, Nantong University, Jiangsu 226001, China
| | - Hao Yang
- Department of Pathophysiology, School of Medicine, Nantong University, Jiangsu 226001, China
| | - Yanhong Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, Jiangsu 226001, China
| | - Yijing Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, Jiangsu 226001, China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, Jiangsu 226001, China..
| |
Collapse
|
21
|
Emmerich PB, Qyli T, Johnson KA, Chaudhuri S, Clark KM, Verhagen NB, Depke MG, Clipson L, Pasch CA, Papadas A, Burkard ME, Wisinski KB, McGregor SM, Asimakopoulos F, Deming DA. Stromal Versican Accumulation and Proteolysis Regulate the Infiltration of CD8 + T Cells in Breast Cancer. Cancers (Basel) 2025; 17:1435. [PMID: 40361362 PMCID: PMC12070914 DOI: 10.3390/cancers17091435] [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: 02/14/2025] [Revised: 03/28/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Recent clinical trials in breast cancer have demonstrated that some patients benefit from immune checkpoint blockade, though better predictive markers are needed. The accumulation of the immunomodulatory matrix proteoglycan versican (VCAN) can predict the exclusion of CD8+ tumor-infiltrating lymphocytes (TILs) in some settings and, thus, is evaluated in breast cancer here. Methods: A total of 230 breast cancers were analyzed for VCAN accumulation, VCAN proteolysis, and CD8+ TILs. CD8+ TILs were categorized based on their localization in the tumor epithelial or stromal compartments. Results: VCAN accumulation was detected in 90% of breast cancers, more commonly in ER+ tumors (93% vs. 77%; p < 0.001). MCF7 cells treated with estrogen upregulate VCAN without an enhanced expression of ADAMTS-proteases. VCAN-undetectable tumors demonstrate greater CD8+ TILs compared to VCAN-detectable tumors (p = 0.012). CD8+ T cells within TNBC tumors with high VCAN proteolysis infiltrated the epithelial compartment more often than in tumors with low VCAN proteolysis (91% vs. 42% respectively; p = 0.008). In the TCGA cohort, a strong inverse correlation between CD8A and VCAN expression was observed across subtypes. Conclusions: VCAN accumulation correlates with the exclusion of CD8+ TILs across subtypes of breast cancer, warranting further validation of VCAN accumulation and proteolysis as predictive biomarkers for breast cancer immunotherapy.
Collapse
Affiliation(s)
- Philip B. Emmerich
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Cellular and Molecular Pathology Graduate Program, Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Tonela Qyli
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Katherine A. Johnson
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Somak Chaudhuri
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kristen M. Clark
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Nathaniel B. Verhagen
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mitchell G. Depke
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Linda Clipson
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Cheri A. Pasch
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Athanasios Papadas
- Cellular and Molecular Pathology Graduate Program, Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Division of Blood and Marrow Transplantation and Moores Cancer Center, University of California-San Diego, La Jolla, CA 92093, USA
| | - Mark E. Burkard
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kari B. Wisinski
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Stephanie M. McGregor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Fotis Asimakopoulos
- Division of Blood and Marrow Transplantation and Moores Cancer Center, University of California-San Diego, La Jolla, CA 92093, USA
| | - Dustin A. Deming
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
22
|
Li G, Yuan Y, Wu X, Wu L. Modulating NPC1L1 to Potentiate PARP Inhibitor-Induced Ferroptosis and Immune Response in Triple-Negative Breast Cancer. Pharmaceutics 2025; 17:554. [PMID: 40430847 PMCID: PMC12114877 DOI: 10.3390/pharmaceutics17050554] [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: 02/17/2025] [Revised: 04/05/2025] [Accepted: 04/16/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Poly (ADP-ribose) polymerase (PARP) inhibitors have shown significant efficacy in treating BRCA-mutated cancers; however, a significant proportion of patients fail to respond. Emerging evidence highlights the role of PARP in lipid metabolism, suggest-ing its modulation as a novel strategy to regulate tumor progression. Methods: In this study, lipidomics and transcriptomics analyses were conducted to elucidate the mechanisms underlying PARP inhibitor-induced ferroptosis and immune modulation in triple-negative breast cancer (TNBC). Results: We demonstrated that the PARP inhibitor Niraparib significantly reprograms lipid metabolism in TNBC cells, marked by elevated phosphatidylethanolamine (PE) and cholesterol ester (ChE) levels. This metabolic shift was mechanistically linked to upregulation of the cholesterol transporter NPC1L1 via the PARP1-RELA-NPC1L1 signaling axis, which subsequently activated the AKT pathway. Combinatorial treatment with Niraparib and either Ezetimibe (an NPC1L1 inhibitor) or AZD5363 (an AKT inhibitor) synergistically enhanced TNBC cell death by promoting ferroptosis through glutathione depletion and lipid peroxidation. Furthermore, NPC1L1 inhibition amplified PARP inhibitor-induced immune responses, increasing CD8+ T cell infiltration and cytotoxicity in tumors. Conclusions: In conclusion, our findings establish NPC1L1 as a critical mediator of PARP inhibitor efficacy and propose dual targeting of lipid metabolism, providing a new therapeutic approach for the combination treatment of TNBC.
Collapse
Affiliation(s)
- Ge Li
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350100, China; (G.L.); (Y.Y.); (X.W.)
- Fujian Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou 350100, China
| | - Yuxia Yuan
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350100, China; (G.L.); (Y.Y.); (X.W.)
- Fujian Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou 350100, China
| | - Xinhua Wu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350100, China; (G.L.); (Y.Y.); (X.W.)
- Fujian Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou 350100, China
| | - Lixian Wu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350100, China; (G.L.); (Y.Y.); (X.W.)
- Fujian Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou 350100, China
| |
Collapse
|
23
|
Mariano NC, Marotti JD, Chen Y, Karakyriakou B, Salgado R, Christensen BC, Miller TW, Kettenbach AN. Quantitative proteomics analysis of triple-negative breast cancers. NPJ Precis Oncol 2025; 9:117. [PMID: 40269124 PMCID: PMC12019170 DOI: 10.1038/s41698-025-00907-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 04/05/2025] [Indexed: 04/25/2025] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for approximately 15% of all Breast Cancer (BC) cases with poorer prognosis and clinical outcomes compared to other BC subtypes due to greater tumor heterogeneity and few therapeutically targetable oncogenic drivers. To reveal actionable pathways for anti-cancer treatment, we use a proteomic approach to quantitatively compare the abundances of 6306 proteins across 55 formalin-fixed and paraffin-embedded (FFPE) TNBC tumors. We identified four major TNBC clusters by unsupervised clustering analysis of protein abundances. Analyses of clinicopathological characteristics revealed associations between the proteomic profiles and clinical phenotypes exhibited by each subtype. We validate the findings by inferring immune and stromal cell type composition from genome-wide DNA methylation profiles. Finally, quantitative proteomics on TNBC cell lines was conducted to identify in vitro models for each subtype. Collectively, our data provide subtype-specific insights into molecular drivers, clinicopathological phenotypes, tumor microenvironment (TME) compositions, and potential pharmacologic vulnerabilities for further investigations.
Collapse
Affiliation(s)
| | - Jonathan D Marotti
- Department of Pathology and Laboratory Medicine, Lebanon, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
| | | | | | - Roberto Salgado
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Brock C Christensen
- Department of Pathology and Laboratory Medicine, Lebanon, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Lebanon, NH, USA
- Department of Epidemiology, Lebanon, NH, USA
- Department of Community and Family Medicine, Lebanon, NH, USA
| | - Todd W Miller
- Dartmouth Cancer Center, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Lebanon, NH, USA
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Hanover, NH, USA.
- Dartmouth Cancer Center, Lebanon, NH, USA.
| |
Collapse
|
24
|
Dos Santos ALS, Da Silva JL, De Albuquerque LZ, Neto ALA, Da Silva CF, Cerva LAM, Small IA, Rodrigues FR, De Macedo FC, Marcelino CP, Batista PDM, Rego MADC, Borba MACSM, De Melo AC. Unveiling the Landscape of PD-L1 Expression and Tumor-Infiltrating Lymphocyte Subtypes in Advanced Triple-Negative Breast Cancer in Brazil. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:349-358. [PMID: 40256247 PMCID: PMC12009053 DOI: 10.2147/bctt.s499373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 03/12/2025] [Indexed: 04/22/2025]
Abstract
Purpose This study aimed to assess the frequency and prognostic significance of programmed cell death ligand 1 (PD-L1) expression and tumor-infiltrating lymphocyte (TIL) subtypes in advanced triple-negative breast cancer (TNBC). Patients and Methods A database search was conducted to identify women with previously untreated locally recurrent inoperable or metastatic TNBC treated between January 2018 and December 2022. The inclusion criteria required formalin-fixed paraffin-embedded samples aged less than four years. PD-L1 expression was evaluated using the PD-L1 IHC 22C3 pharmDx assay, and the combined positive score (CPS) was calculated. TIL subtypes were assessed using immunohistochemical staining. Results The study included 150 patients, with a median age of 51.5 years. The majority of patients were younger than 65 years, postmenopausal, non-white, and had metastatic TNBC. CPS≥10 was observed in 20.9% of cases, mainly in postmenopausal women. No significant differences were found in demographic characteristics and clinicopathological variables across PD-L1 subgroups. Tumors with PD-L1 CPS≥10 had higher expression of CD3+, CD4+, and CD8+ TIL subtypes. Most patients received first-line chemotherapy, with smaller proportions undergoing second, third, and fourth-line treatments. No statistically significant differences were observed in median progression-free survival (PFS) or overall survival (OS) across PD-L1 subgroups in this cohort of chemotherapy-treated patients. Conclusion This study provides insights into the expression profiles of PD-L1 and TIL subtypes in advanced TNBC. The PD-L1 CPS status did not significantly affect survival outcomes, but variations in TIL subtype composition were observed based on PD-L1 CPS status.
Collapse
Affiliation(s)
| | - Jesse Lopes Da Silva
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Lucas Zanetti De Albuquerque
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Antônio Lucas Araújo Neto
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Cecília Ferreira Da Silva
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Luana Aguiar Mesquita Cerva
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Isabele Avila Small
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | - Andreia Cristina De Melo
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| |
Collapse
|
25
|
Li Y, Ding T, Zhang T, Liu S, Wang J, Zhou X, Guo Z, He Q, Zhang S. Leveraging Diverse Cell-Death Patterns to Decipher the Interactive Relation of Unfavorable Outcome and Tumor Microenvironment in Breast Cancer. Bioengineering (Basel) 2025; 12:420. [PMID: 40281780 PMCID: PMC12024675 DOI: 10.3390/bioengineering12040420] [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: 02/19/2025] [Revised: 03/25/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Programmed cell death (PCD) dynamically influences breast cancer (BC) prognosis through interactions with the tumor microenvironment (TME). We investigated 13 PCD patterns to decipher their prognostic impact and mechanistic links to TME-driven outcomes. Our study aimed to explore the complex mechanisms underlying these interactions and establish a prognostic prediction model for breast cancer. METHODS Using TCGA and METABRIC datasets, we integrated single-sample gene set enrichment analysis (ssGSEA), weighted gene co-expression network analysis (WGCNA), and Least Absolute Shrinkage and Selection Operator (LASSO) to explore PCD-TME interactions. Multi-dimensional analyses included immune infiltration, genomic heterogeneity, and functional pathway enrichment. RESULTS Our results indicated that high apoptosis and pyroptosis activity, along with low autophagy, correlated with favorable prognosis, which was driven by enhanced anti-tumor immunity, including more M1 macrophage polarization and activated CD8+ T cells in TME. PCD-related genes could promote tumor metastasis and poor prognosis via VEGF/HIF-1/MAPK signaling and immune response, including Th1/Th2 cell differentiation, while new tumor event occurrences (metastasis/secondary cancers) were linked to specific clinical features and gene mutation spectrums, including TP53/CDH1 mutations and genomic instability. We constructed a six-gene LASSO model (BCAP31, BMF, GLUL, NFKBIA, PARP3, PROM2) to predict prognosis and identify high-risk BC patients (for five-year survival, AUC = 0.76 in TCGA; 0.74 in METABRIC). Therein, the high-risk subtype patients demonstrated a poorer prognosis, also characterized by lower microenvironment matrix and downregulated immunocyte infiltration. These six gene signatures also showed prognostic value with significant differential expression in gene and protein levels of BC samples. CONCLUSION Our study provided a comprehensive landscape of the cancer survival difference and related PCD-TME interaction axis and highlighted that high-apoptosis/pyroptosis states caused favorable prognosis, underlying mechanisms closely related with the TME where anti-tumor immunity would be beneficial for patient prognosis. These findings highlighted the model's potential for risk stratification in BC.
Collapse
Affiliation(s)
- Yue Li
- Department of Clinical Laboratories, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (Y.L.); (T.D.); (Z.G.); (Q.H.)
| | - Ting Ding
- Department of Clinical Laboratories, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (Y.L.); (T.D.); (Z.G.); (Q.H.)
| | - Tong Zhang
- Department of Clinical Laboratories, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (Y.L.); (T.D.); (Z.G.); (Q.H.)
| | - Shuangyu Liu
- Department of Clinical Laboratories, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (Y.L.); (T.D.); (Z.G.); (Q.H.)
| | - Jinhua Wang
- Department of Clinical Laboratories, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (Y.L.); (T.D.); (Z.G.); (Q.H.)
| | - Xiaoyan Zhou
- Department of Clinical Laboratories, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (Y.L.); (T.D.); (Z.G.); (Q.H.)
| | - Zeqi Guo
- Department of Clinical Laboratories, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (Y.L.); (T.D.); (Z.G.); (Q.H.)
| | - Qian He
- Department of Clinical Laboratories, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (Y.L.); (T.D.); (Z.G.); (Q.H.)
| | - Shuqun Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| |
Collapse
|
26
|
Tiwari PK, Chaudhary AA, Gupta S, Chouhan M, Singh HN, Rustagi S, Khan SUD, Kumar S. Extracellular vesicles in triple-negative breast cancer: current updates, challenges and future prospects. Front Mol Biosci 2025; 12:1561464. [PMID: 40297849 PMCID: PMC12034555 DOI: 10.3389/fmolb.2025.1561464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/25/2025] [Indexed: 04/30/2025] Open
Abstract
Breast cancer (BC) remains a complex and widespread problem, affecting millions of women worldwide, Among the various subtypes of BC, triple-negative breast cancer (TNBC) is particularly challenging, representing approximately 20% of all BC cases, and the survival rate of TNBC patients is generally worse than other subtypes of BC. TNBC is a heterogeneous disease characterized by lack of expression of three receptors: estrogen (ER), progesterone (PR), and human epidermal growth factor receptor 2 (HER2), resulting conventional hormonal therapies are ineffective for its management. Despite various therapeutic approaches have been explored, but no definitive solution has been found yet for TNBC. Current treatments options are chemotherapy, immunotherapy, radiotherapy and surgery, although, these therapies have some limitations, such as the development of resistance to anti-cancer drugs, and off-target toxicity, which remain primary obstacles and significant challenges for TNBC. Several findings have shown that EVs exhibit significant therapeutic promise in many diseases, and a similar important role has been observed in various types of tumor. Studies suggest that EVs may offer a potential solution for the management of TNBC. This review highlights the multifaceted roles of EVs in TNBC, emphasizing their involvement in disease progression, diagnosis and therapeutic approach, as well as their potential as biomarkers and drug delivery.
Collapse
Affiliation(s)
- Prashant Kumar Tiwari
- Biological and Bio-Computational Lab, Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Mandeep Chouhan
- Biological and Bio-Computational Lab, Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Himanshu Narayan Singh
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, United States
| | - Sarvesh Rustagi
- Department of Food Technology, School of Applied and Life science, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Salah-Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Sanjay Kumar
- Biological and Bio-Computational Lab, Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
27
|
Chen IP, Henning S, Bender M, Degenhardt S, Mhamdi Ghodbani M, Bergmann AK, Volkmer B, Brockhoff G, Wege AK, Greinert R. Detection of Human Circulating and Extracellular Vesicle-Derived miRNAs in Serum of Humanized Mice Transplanted with Human Breast Cancer (HER2 + and TNBC) Cells-A Proof of Principle Investigation. Int J Mol Sci 2025; 26:3629. [PMID: 40332177 PMCID: PMC12026515 DOI: 10.3390/ijms26083629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Humanized tumor mice (HTM) allow for preclinical cancer treatment studies of breast cancer (BC) under human-like conditions. This study utilized HTM for the first time to investigate potential miRNA biomarker candidates for treatment response in sera and extracellular vesicles (EVs), following X-irradiation and atezolizumab (anti-PD-L1) treatment. We identified the changes of human-specific miRNAs (miR-23b-3p and miR-155-5p) after irradiation and anti-PD-L1 treatment in HTMs with human epidermal growth factor receptor 2 positive (HER2+ BC) and triple-negative breast cancer (TNBC). The high degree of conserved, circulating free miRNA in mice and men represents a challenge of our assay; however, miRNAs with ≥2 nucleotide mismatches can be employed for human-specific analysis, and even conserved miRNAs may be utilized under clearly defined conditions of human tumor growth in HTM. A comparative analysis of extracellular vesicle miRNA cargo and free-circulating serum miRNAs revealed several exosome-specific miRNAs (miR-29b-3p, miR-34c-5p, miR-203a-3p, miR-378g, and miR-382-5p) in HTMs, which are known to play roles in BC. Our findings demonstrate that HTMs are a suitable model to identify treatment-induced changes in free-circulating and exosomal miRNAs that influence tumor progression and immunological tumor defense, both locally and at distant sites. This study presents a proof-of-principle approach to analyzing cell-free nucleotides and exosomes in a human-like, preclinical in vivo setting. Further refinements are necessary to enhance the sensitivity and the specificity of the HTM-based approach.
Collapse
Affiliation(s)
- I-Peng Chen
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Stefan Henning
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Marc Bender
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Sarah Degenhardt
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Mouna Mhamdi Ghodbani
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Ann Kathrin Bergmann
- Core Facility of Electron Microscopy, University Clinics Duesseldorf, 40225 Duesseldorf, Germany;
| | - Beate Volkmer
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Gero Brockhoff
- Department of Gynecology and Obstetrics, Medical Center Regensburg, 93053 Regensburg, Germany; (G.B.); (A.K.W.)
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| | - Anja K. Wege
- Department of Gynecology and Obstetrics, Medical Center Regensburg, 93053 Regensburg, Germany; (G.B.); (A.K.W.)
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| | - Rüdiger Greinert
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| |
Collapse
|
28
|
Srisawat W, Koonyosying P, Muenthaisong A, Sangkakam K, Varinrak T, Rittipornlertrak A, Nambooppha B, Apinda N, Sthitmatee N. mRNA and protein expression of programmed cell death-ligand-1 on canine mammary gland tumour in dogs of Chiang Mai, Thailand. Int J Vet Sci Med 2025; 13:1-11. [PMID: 40206791 PMCID: PMC11980185 DOI: 10.1080/23144599.2025.2483102] [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: 12/05/2024] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 04/11/2025] Open
Abstract
Metastasis-related disease is a major cause of death in canine mammary tumours (CMTs). Immunotherapy has been investigated due to the less successful outcomes of systemic therapy. This study aims to examine the expression of Programmed Cell Death Ligand-1 (PD-L1) in canine mammary tumours in dogs of Chiang Mai, Thailand, and determine the relationship between the level of mRNA expression and clinicopathologic characteristics. A total of 28 CMT samples were collected at the Small Animal Hospital, Chiang Mai University. Quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) and western blot assays were performed. The results revealed that all CMTs in this study expressed PD-L1 mRNA and PD-L1 protein. The mean relative mRNA expression showed no significant differences between groups categorized by age, tumour size, or histopathological findings. However, the mean relative mRNA expression in tumours with a TNM stage >3 was significantly lower compared to those with TNM stage ≤2. In conclusion, this study investigates the expression of PD-L1 mRNA and PD-L1 protein, particularly in malignant CMTs. The findings strongly support the potential for developing effective immunotherapy methods targeting the PD-1/PD-L1 pathway for advanced CMTs in the future. For further conclusive assessment, future studies should focus on refining immunotherapy strategies for CMT cases expressing PD-L1.
Collapse
Affiliation(s)
- Wanwisa Srisawat
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai, Thailand
| | - Pongpisid Koonyosying
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai, Thailand
| | - Anucha Muenthaisong
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai, Thailand
| | - Kanokwan Sangkakam
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thanya Varinrak
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Veterinary Medical Diagnostic and Animal Health Innovation, Chiang Mai University, Chiang Mai, Thailand
| | - Amarin Rittipornlertrak
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Boondarika Nambooppha
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nisachon Apinda
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nattawooti Sthitmatee
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Research Center for Veterinary Bioscience and Veterinary Public Health, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
29
|
Shen J, Ye X, Hou H, Wang Y. Efficacy and Safety of Immunochemotherapy in Advanced Triple-negative Breast Cancer: A Meta-analysis of Randomised Clinical Trials. Clin Oncol (R Coll Radiol) 2025; 40:103783. [PMID: 39955967 DOI: 10.1016/j.clon.2025.103783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/19/2024] [Accepted: 01/30/2025] [Indexed: 02/18/2025]
Abstract
AIMS Based on the existing controversial clinical research results, this study conducted a comprehensive meta-analysis of relevant literature to clarify the benefits of immunochemotherapy (ICT)-which combines immune checkpoint inhibitors and chemotherapy (CT)-for patients with advanced triple-negative breast cancer (aTNBC). MATERILAS AND METHODS A thorough literature search was conducted up to February 15, 2024. Subsequently, meta-analyses were performed to aggregate hazard ratios (HRs) for progression-free survival (PFS) and overall survival (OS), odds ratios (ORs) for objective response rate (ORR) and relative risks (RRs) for adverse events (AEs). RESULTS Six randomised clinical trials (RCTs) involving 3,105 patients met the inclusion criteria. In comparison with CT, ICT yielded significant enhancements in PFS (HR, 0.80; 95%CI: 0.73-0.87), OS (HR, 0.87; 95%CI: 0.80-0.96), and ORR (OR, 1.34; 95%CI: 1.15-1.55) in the intention-to-treat population. However, ICT also exhibited an increase in grade ≥3 AEs (RR, 1.11; 95%CI: 1.04-1.19) and severe AEs (RR, 1.40; 95%CI: 1.18-1.66). Subgroup analyses revealed that ICT significantly improved PFS (HR, 0.67; 95%CI: 0.58-0.77), OS (HR, 0.75; 95%CI: 0.64-0.87), and ORR (OR, 1.47; 95%CI: 1.16-1.84) within the PD-L1-positive subgroup, whereas no statistically significant differences were detected for PD-L1-negative population. CONCLUSION ICT demonstrates superior efficacy over conventional CT in the treatment of aTNBC, albeit accompanied by heightened toxicity. Notably, the assessment of PD-L1 status may serve as a valuable biomarker in discerning aTNBC patients who are particularly predisposed to derive benefit from ICT. PROSPERO NUMBER CRD42024513270.
Collapse
Affiliation(s)
- J Shen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - X Ye
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - H Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China.
| | - Y Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China.
| |
Collapse
|
30
|
Huang J, Shi J, Ma N, Li Y, Jin W, Zhang H, Zhang X, Luo N, Ding Y, Xie Q, Li Q, Xiong Y. Celastrol-loaded ginsenoside Rg3 liposomes enhance anti-programmed death ligand 1 immunotherapy by inducing immunogenic cell death in triple-negative breast cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156514. [PMID: 39986227 DOI: 10.1016/j.phymed.2025.156514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC), characterized by high heterogeneity and invasiveness. Currently, inducing immunogenic cell death (ICD) of tumor cells through approaches such as radiotherapy and chemotherapy is an effective strategy to enhance the response to anti-programmed death-ligand 1 antibody (aPD-L1) therapy in TNBC. However, radiotherapy and chemotherapy treatments often upregulate PD-L1 expression in tumor cells, thereby weakening the tumor cells' response to aPD-L1. Celastrol exhibits broad-spectrum and potent anti-tumor activity, efficiently inducing ICD without increasing PD-L1 levels in tumor cells. PURPOSE This study aims to elucidate the tumor-targeting effects of celastrol-loaded liposomes and its synergistic efficacy and mechanism of action in combination with aPD-L1 against TNBC. METHODS The Rg3 liposomes loaded with celastrol (Cel-Rg3-Lp) were prepared using the thin-film hydration method. BALB/c mice were utilized to establish an in situ breast cancer model. Mice were intravenously injected with Cel-Rg3-Lp at a dosage of celastrol 1 mg/kg once every two days for a total of 7 injections. Flow cytometry, western blot, and immunofluorescence techniques were employed to investigate the synergistic effects and mechanisms of Cel-Rg3-Lp combined with aPD-L1 in the treatment of TNBC. RESULTS The findings of this study demonstrate that after 7 administrations of Cel-Rg3-Lp (1 mg/kg celastrol, intravenously), significant anti-tumor effects are observed, including the recruitment of CD8+T cells and dendritic cells (DCs), while reducing the infiltration of immunosuppressive cells. The therapeutic efficacy was further enhanced when combined with aPD-L1. Additionally, Cel-Rg3-Lp markedly downregulated glucose-regulated protein 78 (GRP78) expression, thereby inducing ICD in tumor cells. CONCLUSION This study successfully constructed a multifunctional liposome and proposed a mechanism for inducing ICD through the GRP78-endoplasmic reticulum stress pathway. The liposome downregulates GRP78, triggering endoplasmic reticulum stress in tumor cells, inducing ICD, activating DCs, and enhancing antigen presentation to T cells. This improves the tumor immune microenvironment and provides a theoretical foundation for combining Cel-Rg3-Lp with aPD-L1 in the treatment of TNBC. This mechanism opens unique prospects for using celastrol in TNBC therapy and enhancing the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Jingyi Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jingbin Shi
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ninghui Ma
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yujie Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Wanyu Jin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Hongyan Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Xin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ningchao Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ye Ding
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Qiong Xie
- Gynecology Department, Zhoushan Hospital of Traditional Chinese Medicine (Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University), Zhoushan, Zhejiang 316000, China.
| | - Qiushuang Li
- Center of Clinical Evaluation and Analysis, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310053, China.
| | - Yang Xiong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
31
|
Palma M. Advancing Breast Cancer Treatment: The Role of Immunotherapy and Cancer Vaccines in Overcoming Therapeutic Challenges. Vaccines (Basel) 2025; 13:344. [PMID: 40333213 PMCID: PMC12030785 DOI: 10.3390/vaccines13040344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 05/09/2025] Open
Abstract
Breast cancer (BC) remains a significant global health challenge due to its complex biology, which complicates both diagnosis and treatment. Immunotherapy and cancer vaccines have emerged as promising alternatives, harnessing the body's immune system to precisely target and eliminate cancer cells. However, several key factors influence the selection and effectiveness of these therapies, including BC subtype, tumor mutational burden (TMB), tumor-infiltrating lymphocytes (TILs), PD-L1 expression, HER2 resistance, and the tumor microenvironment (TME). BC subtypes play a critical role in shaping treatment responses. Triple-negative breast cancer (TNBC) exhibits the highest sensitivity to immunotherapy, while HER2-positive and hormone receptor-positive (HR+) subtypes often require combination strategies for optimal outcomes. High TMB enhances immune responses by generating neoantigens, making tumors more susceptible to immune checkpoint inhibitors (ICIs); whereas, low TMB may indicate resistance. Similarly, elevated TIL levels are associated with better immunotherapy efficacy, while PD-L1 expression serves as a key predictor of checkpoint inhibitor success. Meanwhile, HER2 resistance and an immunosuppressive TME contribute to immune evasion, highlighting the need for multi-faceted treatment approaches. Current breast cancer immunotherapies encompass a range of targeted treatments. HER2-directed therapies, such as trastuzumab and pertuzumab, block HER2 dimerization and enhance antibody-dependent cellular cytotoxicity (ADCC), while small-molecule inhibitors, like lapatinib and tucatinib, suppress HER2 signaling to curb tumor growth. Antibody-drug conjugates (ADCs) improve tumor targeting by coupling monoclonal antibodies with cytotoxic agents, minimizing off-target effects. Meanwhile, ICIs, including pembrolizumab, restore T-cell function, and CAR-macrophage (CAR-M) therapy leverages macrophages to reshape the TME and overcome immunotherapy resistance. While immunotherapy, particularly in TNBC, has demonstrated promise by eliciting durable immune responses, its efficacy varies across subtypes. Challenges such as immune-related adverse events, resistance mechanisms, high costs, and delayed responses remain barriers to widespread success. Breast cancer vaccines-including protein-based, whole-cell, mRNA, dendritic cell, and epitope-based vaccines-aim to stimulate tumor-specific immunity. Though clinical success has been limited, ongoing research is refining vaccine formulations, integrating combination therapies, and identifying biomarkers for improved patient stratification. Future advancements in BC treatment will depend on optimizing immunotherapy through biomarker-driven approaches, addressing tumor heterogeneity, and developing innovative combination therapies to overcome resistance. By leveraging these strategies, researchers aim to enhance treatment efficacy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Marco Palma
- Institute for Globally Distributed Open Research and Education (IGDORE), 03181 Torrevieja, Spain
| |
Collapse
|
32
|
Su Y, Bai Q, Zhang W, Xu B, Hu T. The Role of Long Non-Coding RNAs in Modulating the Immune Microenvironment of Triple-Negative Breast Cancer: Mechanistic Insights and Therapeutic Potential. Biomolecules 2025; 15:454. [PMID: 40149989 PMCID: PMC11939868 DOI: 10.3390/biom15030454] [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: 02/18/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous and aggressive subtype of breast cancer that faces therapeutic challenges due to a shortage of effective targeted therapies. The complex biology of TNBC renders its clinical management fraught with difficulties, especially regarding the immune microenvironment of the tumor. In recent years, long non-coding RNAs (lncRNAs) have been recognized as important gene regulators with key roles in tumor development and microenvironmental regulation. Previous studies have shown that lncRNAs play important roles in the immune microenvironment of TNBC, including the regulation of tumor immune escape and the function of tumor-infiltrating immune cells. However, despite the increasing research on lncRNAs, there are still many unanswered questions, such as their specific mechanism of action and how to effectively utilize them as therapeutic targets. Therefore, the aim of this study was to review the mechanisms of lncRNAs in the TNBC immune microenvironment, explore their regulatory roles in tumor immune escape and immune cell infiltration, and explore their prospects as potential therapeutic targets. By integrating the latest research results, this study aims to provide new ideas and directions for future TNBC treatment.
Collapse
Affiliation(s)
- Yongcheng Su
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (Q.B.); (W.Z.)
| | - Qingquan Bai
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (Q.B.); (W.Z.)
| | - Wenqing Zhang
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (Q.B.); (W.Z.)
| | - Beibei Xu
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Tianhui Hu
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (Q.B.); (W.Z.)
- Shenzhen Research Institute, Xiamen University, Shenzhen 518057, China
| |
Collapse
|
33
|
Lin Y, Song Y, Zhang Y, Li X, Kan L, Han S. New insights on anti-tumor immunity of CD8 + T cells: cancer stem cells, tumor immune microenvironment and immunotherapy. J Transl Med 2025; 23:341. [PMID: 40097979 PMCID: PMC11912710 DOI: 10.1186/s12967-025-06291-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/23/2025] [Indexed: 03/19/2025] Open
Abstract
Recent breakthroughs in tumor immunotherapy have confirmed the capacity of the immune system to fight several cancers. The effective means of treating cancer involves accelerating the death of tumor cells and improving patient immunity. Dynamic changes in the tumor immune microenvironment alter the actual effects of anti-tumor drug production and may trigger favorable or unfavorable immune responses by modulating tumor-infiltrating lymphocytes. Notably, CD8+ T cells are one of the primary tumor-infiltrating immune cells that provide anti-tumor response. Tumor cells and tumor stem cells will resist or evade destruction through various mechanisms as CD8+ T cells exert their anti-tumor function. This paper reviews the research on the regulation of tumor development and prognosis by cancer stem cells that directly or indirectly alter the role of tumor-infiltrating CD8+ T cells. We also discuss related immunotherapy strategies.
Collapse
Affiliation(s)
- Yibin Lin
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yifu Song
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yaochuan Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaodong Li
- Department of Neurosurgery, Siping Central People's Hospital, Siping, Jilin, 136000, China
| | - Liang Kan
- Department of Geriatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Sheng Han
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
34
|
Jin L, Yang Z, Tang W, Yu P, Chen R, Xu Y, Zhang J. The evolving landscape of genetic biomarkers for immunotherapy in primary and metastatic breast cancer. Front Oncol 2025; 15:1522262. [PMID: 40182039 PMCID: PMC11966456 DOI: 10.3389/fonc.2025.1522262] [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: 11/04/2024] [Accepted: 01/30/2025] [Indexed: 04/05/2025] Open
Abstract
Background Major advances have been achieved in the characterization of primary breast cancer genomic profiles. Limited information is available on the genomic profile of tumors originating from different metastatic locations in recurrent/metastatic (R/M) breast cancer, especially in Asian patients. This study aims to decipher the mutational profiles of primary and R/M breast cancer in Chinese patients using next-generation sequencing. Methods A total of 563 breast cancer patients were enrolled, and 590 tumor tissues and matched peripheral blood samples were collected and subjected to targeted sequencing with a panel of 1,021 cancer-related genes. The mutation spectrum, DNA damage response (DDR) genes, commonly altered signal pathways, and immunotherapy-related markers were compared between primary and R/M breast cancer. The molecular differences between our cohort and the Memorial Sloan Kettering Cancer Center (MSKCC) dataset were also explored. Results A total of 361 samples from primary and 229 samples from R/M breast cancer were analyzed. BRCA2, ATRX, and ATM were more frequently observed in R/M lesions among the 36 DDR genes. An ESR1 mutation and PD-L1 and PD-L2 amplification were enriched in R/M breast cancer (all p<0.05). Compared with the MSKCC dataset, we recruited more patients diagnosed at age 50 or younger and more patients with triple-negative breast cancer (TNBC) subtypes. The TNBC patients in our dataset had a higher percentage of PD-L1 amplification in metastasis tumors (p<0.05). Conclusions This study revealed the distinctive mutational features of primary and R/M tumors in Chinese breast cancer patients, which are different from those from Western countries. The enrichment of PD-L1 amplification in metastatic TNBC indicates the necessity to re-biopsy metastatic tumors for immunotherapy.
Collapse
Affiliation(s)
- Liang Jin
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zijian Yang
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Wei Tang
- Department of Breast Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pengli Yu
- Medical Department, Geneplus-Beijing, Beijing, China
| | - Rongrong Chen
- Medical Department, Geneplus-Beijing, Beijing, China
| | - Yan Xu
- Department of Breast and Thyroid Surgery, Daping Hospital, Army Military Medical University, Chongqing, China
| | - Jun Zhang
- Department of Thyroid and Breast Surgery, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| |
Collapse
|
35
|
Sun Z, Liu C, Yao Y, Gao C, Li H, Wang L, Li Y, Sun C. Therapeutic potential of triple-negative breast cancer immune checkpoint blockers: A 21-year bibliometric analysis. Medicine (Baltimore) 2025; 104:e41739. [PMID: 40068043 PMCID: PMC11903016 DOI: 10.1097/md.0000000000041739] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive metastatic subtype of BC that frequently develops chemoresistance. Immune checkpoint blockers (ICB) have led to breakthroughs in TNBC treatment. This study aimed to explore research trends and public interest in ICB interventions for TNBC. METHODS We searched the Web of Science Core Collection (WoSCC) database for publications related to ICB for TNBC from 2003 to 2024. VOSviewer, CiteSpace, and R package "bibliometrix" were used to analyze the characteristics of ICB publications in TNBC from a quantitative and qualitative perspective and to visualize the results to comprehensively present the research trends in this field. RESULTS After removing duplicates, 2698 publications were included. The New England Journal of Medicine may be the leading and influential in the field of ICB in TNBC according to data on the total number of publications, number of citations, and impact factors. Its article entitled "Atezolizumab and Nab-Paclitaxel in Advanced TNBC" is 1 of the most cited articles. Keyword analysis showed that current research hotspots in this field are tumor microenvironment, complete pathological response, neoadjuvant chemotherapy, and PARP inhibitors. Future research hotspots may include the PD-L1 inhibitor durvalumab and antibody-drug conjugates (ADC). CONCLUSIONS This study revealed that ICB therapy for TNBC is a rapidly evolving and high-profile topic. Future research should focus on the optimal selection of different targets for ICB in combination with neoadjuvant chemotherapy, ADC, and poly ADP-ribose polymerase inhibitors to treat TNBC.
Collapse
Affiliation(s)
- Zhongli Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Traditional Chinese Medicine, Chongqing Three Gorges Medical College, Chongqing, China
| | - Cun Liu
- Department of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Yan Yao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chundi Gao
- Department of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Huayao Li
- Department of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Longyun Wang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Ye Li
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Changgang Sun
- Department of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China,
| |
Collapse
|
36
|
Carbone FP, Ancona P, Volinia S, Terrazzan A, Bianchi N. Druggable Molecular Networks in BRCA1/BRCA2-Mutated Breast Cancer. BIOLOGY 2025; 14:253. [PMID: 40136510 PMCID: PMC11940086 DOI: 10.3390/biology14030253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025]
Abstract
Mutations in the tumor suppressor genes BRCA1 and BRCA2 are associated with the triple-negative breast cancer phenotype, particularly aggressive and hard-to-treat tumors lacking estrogen, progesterone, and human epidermal growth factor receptor 2. This research aimed to understand the metabolic and genetic links behind BRCA1 and BRCA2 mutations and investigate their relationship with effective therapies. Using the Cytoscape software, two networks were generated through a bibliographic analysis of articles retrieved from the PubMed-NCBI database. We identified 98 genes deregulated by BRCA mutations, and 24 were modulated by therapies. In particular, BIRC5, SIRT1, MYC, EZH2, and CSN2 are influenced by BRCA1, while BCL2, BAX, and BRIP1 are influenced by BRCA2 mutation. Moreover, the study evaluated the efficacy of several promising therapies, targeting only BRCA1/BRCA2-mutated cells. In this context, CDDO-Imidazolide was shown to increase ROS levels and induce DNA damage. Similarly, resveratrol decreased the expression of the anti-apoptotic gene BIRC5 while it increased SIRT1 both in vitro and in vivo. Other specific drugs were found to induce apoptosis selectively in BRCA-mutated cells or block cell growth when the mutation occurs, i.e., 3-deazaneplanocin A, genistein or daidzein, and PARP inhibitors. Finally, over-representation analysis on the genes highlights ferroptosis and proteoglycan pathways as potential drug targets for more effective treatments.
Collapse
Affiliation(s)
- Francesca Pia Carbone
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
| | - Pietro Ancona
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
| | - Stefano Volinia
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
- Genomics Core Facility, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
- Laboratory for Technologies of Advanced Therapies (LTTA), 44121 Ferrara, Italy
| | - Anna Terrazzan
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
- Genomics Core Facility, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
- Laboratory for Technologies of Advanced Therapies (LTTA), 44121 Ferrara, Italy
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
| |
Collapse
|
37
|
Andour L, Hagenaars SC, Gregus B, Tőkes AM, Karancsi Z, Tollenaar RAEM, Kroep JR, Kulka J, Mesker WE. The prognostic value of the tumor-stroma ratio compared to tumor-infiltrating lymphocytes in triple-negative breast cancer: a review. Virchows Arch 2025; 486:427-444. [PMID: 39904885 PMCID: PMC11950021 DOI: 10.1007/s00428-025-04039-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/17/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
Previous literature extensively explored biomarkers to personalize treatment for breast cancer patients. The clinical need is especially high in patients with triple-negative breast cancer (TNBC) due to its aggressive nature and limited treatment modalities. This review aims to evaluate the value of tumor-infiltrating lymphocytes (TILs) and tumor-stroma ratio (TSR) as prognostic biomarkers in TNBC patients and assess their clinical potential. A literature search was conducted in PubMed, Embase, Emcare, Web of Science, and Cochrane Library. Papers comparing survival outcomes of TNBC patients with low/high or negative/positive TSR and immune cells were included. The most frequently mentioned subgroups of TILs were selected and reported in this review. Data from 43 articles on TILs and eight articles on TSR were included. Among TNBC patients, high CD8 expression was generally associated with better survival. Notable, the poor survival outcomes were related to high intra-tumoral PD-L1 expression, whereas high stromal PD-L1 expression more often was correlated with favorable outcomes. For the TSR, a high amount of stroma in the primary tumor of TNBC patients was consistently associated with worse survival. This review highlights that a high number of CD8-positive T-cells is a promising prognostic factor for TNBC patients. PD-L1 expression analyzed for intra-tumoral and stromal expression separately reports strong but contrasting information. Finally, the TSR shows potential to be an important prognostic marker, especially for TNBC patients. Utilizing both biomarkers, either on itself or combined, could enhance clinical decision-making and personalization of treatment.
Collapse
Affiliation(s)
- Layla Andour
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Sophie C Hagenaars
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Barbara Gregus
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Mária Tőkes
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Zsófia Karancsi
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Rob A E M Tollenaar
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Janina Kulka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Wilma E Mesker
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
38
|
Fan J, Qin Y, Qiu W, Liang J, Xiao C, Xie Q, Tong C, Yuan L, Long Y, Liu B. Gamabufotalin loaded micro-nanocomposites for multimodal therapy of metastatic TNBC by efficiently inducing ICD. Biomaterials 2025; 314:122851. [PMID: 39366186 DOI: 10.1016/j.biomaterials.2024.122851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/30/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Gamabufotalin (CS-6), a main active compound derived from Chinese medicine Chansu, exhibits a robust inhibitory effect on programmed death-ligand 1 (PD-L1) in triple-negative breast cancer (TNBC) cells. Despite its potential for tumor therapy, the medical application of CS-6 is constrained by its hydrophobic nature, lack of targeting capability, and weak immunogenic cell death (ICD) effect. To address these limitations and improve the therapeutic efficiency of this drug against metastatic TNBC, we designed a new kind of CS-6@CPB-S.lux that integrates carboxy-Prussian blue nanoparticles (CPB NPs), CS-6, and attenuated Salmonella typhimurium (S.lux) for TNBC therapy. In vitro and in vivo results have confirmed that CS-6@CPB NPs were efficiently delivered to neoplastic tissue by the tumor hypoxic chemotaxis property of S.lux, wherein the nanomedicine induced significant tumor cell necroptosis and apoptosis via photothermal therapy (PTT) of CPB NPs and chemotherapy of CS-6, which elicited ICD and inhibited PD-L1 expression, resulting in dendritic cells (DCs) maturation and effector T cells activation to comprehensively eliminate tumors. Additionally, the CS-6@CPB-S.lux + Laser treatment significantly transformed the immunosuppressive tumor microenvironment (TME), enhancing antitumor immunity through promoting the polarization of tumor-associated macrophages into antitumorigenic M1 and reducing Tregs recruitment. Consequently, this comprehensive therapy not only inhibited primary and abscopal tumor progression but also prevented TNBC metastasis, which significantly prolonged survival time in animal models. In summary, these findings indicated an alternative approach for metastatic TNBC therapy.
Collapse
Affiliation(s)
- Jialong Fan
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Yan Qin
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China; TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Wensheng Qiu
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Jiahao Liang
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Chang Xiao
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Qian Xie
- Department of Pharmacy, Maternal and Child Health of Hunan Province, Changsha, 410008, China
| | - Chunyi Tong
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Liqin Yuan
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Ying Long
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China.
| | - Bin Liu
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China; NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
39
|
Goerdt L, Stefanovic A, Wirtz R, Karic U, Kohler M, Deutsch TM, Schneeweiss A, Sütterlin M, Stefanovic S, Hofmann J, Wallwiener M. Correlation of HLA-A and HLA-B/C Expression With PD-1 and PD-L1 Expression in Patients With Metastatic Breast Cancer as a Potential Prognosticator of Favorable Survival. In Vivo 2025; 39:758-765. [PMID: 40010968 PMCID: PMC11884484 DOI: 10.21873/invivo.13880] [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: 12/22/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND/AIM Class 1 human leukocyte antigen (HLA) ensures that cytotoxic T lymphocytes (CTLs) attack tumor cells. As part of tumor de-differentiation, the expression of HLA on the tumor cell surface may decrease, which can facilitate tumor growth. Therefore, reduced expression of HLA is generally negatively associated with overall survival (OS). The reverse is true for programmed cell death protein (PD-1) and programmed death ligand 1 (PD-L1). The presence of PD-1 and PD-L1 on the surface of cancer cells inhibits immune defense mechanisms against cancer cells. Therefore, one would expect that increased PD-1/PDL-1 expression would result in decreased 5-year survival. The aim of this study was to correlate the expression levels of HLA-A and HLA-B/C with the expression levels of PD-1 and PDL-1 to evaluate the reliability of their prediction of 5-year OS. MATERIALS AND METHODS This study retrospectively examined patients upon the start of a new therapy line for metastatic breast cancer (MBC). The pilot cohort fulfilling very demanding inclusion criteria consisted of 34 patients. The diagnostics were primarily carried out using RT-qPCR. RESULTS The expression of HLA-A, HLA-B/C, PD-1, and PD-L1 is not significantly associated with OS. CONCLUSION This pilot study found no significant association between HLA-A, HLA-B/C, PD-1, or PD-L1 expression and OS in MBC, indicating limited prognostic value for these biomarkers in this cohort.
Collapse
Affiliation(s)
- Lukas Goerdt
- Department of Gynecology and Obstetrics, Mannheim University Hospital, Heidelberg University, Mannheim, Germany
| | - Aleksandra Stefanovic
- Department of Gynecology and Obstetrics, Mannheim University Hospital, Heidelberg University, Mannheim, Germany
- Brüderklinikum Julia Lanz - Diakonissenkrankenhaus, Mannheim, Germany
- IMDI Science Center, Belgrade, Serbia
| | - Ralph Wirtz
- Stratifyer Molecular Pathology GmbH, Cologne, Germany
| | - Uros Karic
- IMDI Science Center, Belgrade, Serbia
- Hospital for Infectious and Tropical Diseases, Belgrade University School of Medicine, Belgrade, Serbia
| | - Maximilian Kohler
- Department of Gynecology and Obstetrics, Mannheim University Hospital, Heidelberg University, Mannheim, Germany
| | - Thomas M Deutsch
- Department of Gynecology and Obstetrics, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases, Department of Medical Oncology, Heidelberg University, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marc Sütterlin
- Department of Gynecology and Obstetrics, Mannheim University Hospital, Heidelberg University, Mannheim, Germany
| | - Stefan Stefanovic
- Department of Gynecology and Obstetrics, Mannheim University Hospital, Heidelberg University, Mannheim, Germany;
- IMDI Science Center, Belgrade, Serbia
| | - Jan Hofmann
- Department of Gynecology and Obstetrics, Mannheim University Hospital, Heidelberg University, Mannheim, Germany
| | - Markus Wallwiener
- Department of Gynecology, Halle University Hospital, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
40
|
Chmelyuk N, Kordyukova M, Sorokina M, Sinyavskiy S, Meshcheryakova V, Belousov V, Abakumova T. Inhibition of Thioredoxin-Reductase by Auranofin as a Pro-Oxidant Anticancer Strategy for Glioblastoma: In Vitro and In Vivo Studies. Int J Mol Sci 2025; 26:2084. [PMID: 40076706 PMCID: PMC11900239 DOI: 10.3390/ijms26052084] [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: 01/14/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Reactive oxygen species (ROS) play a key role in cancer progression and antitumor therapy. Glioblastoma is a highly heterogeneous tumor with different cell populations exhibiting various redox statuses. Elevated ROS levels in cancer cells promote tumor growth and simultaneously make them more sensitive to anticancer drugs, but further elevation leads to cell death and apoptosis. Meanwhile, various subsets of tumor cells, such a glioblastoma stem cells (GSC) or the cells in tumor microenvironment (TME), demonstrate adaptive mechanisms to excessive ROS production by developing effective antioxidant systems such as glutathione- and thioredoxin-dependent. GSCs demonstrate higher chemoresistance and lower ROS levels than other glioma cells, while TME cells create a pro-oxidative environment and have immunosuppressive effects. Both subpopulations have become an attractive target for developing therapies. Increased expression of thioredoxin reductase (TrxR) is often associated with tumor progression and poor patient survival. Various TrxR inhibitors have been investigated as potential anticancer therapies, including nitrosoureas, flavonoids and metallic complexes. Gold derivatives are irreversible inhibitors of TrxR. Among them, auranofin (AF), a selective TrxR inhibitor, has proven its effectiveness as a drug for the treatment of rheumatoid arthritis and its efficacy as an anticancer agent has been demonstrated in preclinical studies in vitro and in vivo. However, further clinical application of AF could be challenging due to the low solubility and insufficient delivery to glioblastoma. Different delivery strategies for hydrophobic drugs could be used to increase the concentration of AF in the brain. Combining different therapeutic approaches that affect the redox status of various glioma cell populations could become a new strategy for treating brain tumor diseases.
Collapse
Affiliation(s)
- Nelly Chmelyuk
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Laboratory of Biomedical nanomaterials, National Research Technological University “MISIS”, Leninskiy Prospekt 4, 119049 Moscow, Russia
| | - Maria Kordyukova
- Neurotechnology Laboratory, Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117513 Moscow, Russia
| | - Maria Sorokina
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Neurotechnology Laboratory, Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117513 Moscow, Russia
| | - Semyon Sinyavskiy
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Valeriya Meshcheryakova
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Vsevolod Belousov
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Neurotechnology Laboratory, Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117513 Moscow, Russia
| | - Tatiana Abakumova
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
41
|
Zhou B, Chen M, Hao Z, Li L, Zhang Y, Fang B, Shao M, Ren G, Wang K, Liu H, Zhu J, Zhang X, Yuan S, Sitou I, Zhao J, Huang J, Yu Z, Qiu F. Zinc-copper bimetallic nanoplatforms trigger photothermal-amplified cuproptosis and cGAS-STING activation for enhancing triple-negative breast cancer immunotherapy. J Nanobiotechnology 2025; 23:137. [PMID: 39994712 PMCID: PMC11849371 DOI: 10.1186/s12951-025-03186-4] [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: 11/25/2024] [Accepted: 02/01/2025] [Indexed: 02/26/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by high rates of metastasis and recurrence, along with a low sensitivity to immunotherapy, resulting in a paucity of effective therapeutic strategies. Herein, we have developed polydopamine-coated zinc-copper bimetallic nanoplatforms (Cu-ZnO2@PDA nanoplatforms, abbreviated CZP NPs) that can efficiently induce photothermal amplified cuproptosis and cGAS-STING signaling pathway activation, thereby reversing the immunosuppressive tumor microenvironment of TNBC, upregulating PD-L1 expression, and boosting the efficacy of anti-programmed death-ligand 1 antibody (αPD-L1)-based immunotherapy. Within the acidic tumor microenvironment (TME), CZP NPs spontaneously release copper and zinc ions and hydrogen peroxide, generating highly oxidative hydroxyl radicals and downregulating iron-sulfur cluster proteins. These actions lead to the disruption of mitochondrial integrity, the release of mitochondrial DNA (mtDNA) and irreversible cuproptosis. The further synergy between mtDNA and zinc ions potentiates the activation of the cGAS-STING signaling pathway, triggering a robust antitumor immune response and sensitizing TNBC to αPD-L1 therapy. Additionally, using an 808 nm near-infrared laser for photothermal therapy significantly augments these effects, resulting in a cascade amplification of therapeutic efficacy against TNBC. The strategic combination of CZP NPs with αPD-L1 markedly bolsters antitumor immunity and suppresses tumor growth. Collectively, our findings present a promising synergistic strategy for TNBC treatment by linking cuproptosis, cGAS-STING activation, photothermal therapy, and immunotherapy.
Collapse
Affiliation(s)
- Bangyi Zhou
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Mengyao Chen
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Zhixing Hao
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Lili Li
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Yixin Zhang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Baoru Fang
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, P. R. China
| | - Miner Shao
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Guohong Ren
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Ke Wang
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, P. R. China
| | - Huiying Liu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Jingxuan Zhu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Xinyi Zhang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Shuyan Yuan
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - I Sitou
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Jing Zhao
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Jian Huang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
| | - Zhangsen Yu
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, P. R. China.
| | - Fuming Qiu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
| |
Collapse
|
42
|
Yang J, Chen M, Li R, Sun Y, Ye P, Fang K, Wang C, Shi S, Dong C. A responsive cocktail nano-strategy breaking the immune excluded state enhances immunotherapy for triple negative breast cancer. NANOSCALE 2025; 17:4610-4623. [PMID: 39810651 DOI: 10.1039/d4nr03054k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The exclusion of immune cells from the tumor can limit the effectiveness of immunotherapy in triple negative breast cancer (TNBC). The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway plays a crucial role in priming adaptive anti-tumor immunity through the production of type I interferons (IFNs), facilitating the maturation of dendritic cells (DCs) and the function of T cells. Although the increased expression of programmed death-ligand 1 (PD-L1) upon STING activation is favorable for amplifying the efficacy of immune checkpoint inhibitors (ICIs) and realizing combination therapy, the penetration barrier remains a major obstacle. Herein, we fabricated a smart-responsive nanosystem (B&V@ZB-MCL) by integrating the extracellular matrix (ECM)-degrading drug losartan with a STING agonist (Vadimezan, abbreviated to Vad) and a PD-L1 inhibitor (BMS-1). Losartan was first released in the acidic tumor microenvironment to overcome the physical barrier, enhancing the penetration of immunotherapeutic components. Under the triggering of 1O2 generated by a photosensitizer (Ce6), the reactive oxygen species (ROS)-sensitive degradation of the nanocore ensured the site-directed release of Vad and BMS-1. The released Vad and damaged tumor DNA activated immune responses through the cGAS-STING pathway, while the elevated expression level of PD-L1 promoted the anti-tumor effect of BMS-1. Significant degradation of collagen fibers, restoration of immune effector cells, and lower tumor volume were observed in this integrated triple drug sequential therapy, which provides a promising prospect for TNBC treatment.
Collapse
Affiliation(s)
- Jingxian Yang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Mengyao Chen
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Ruihao Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Yanting Sun
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Pingting Ye
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Kang Fang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Chunhui Wang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Shuo Shi
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Chunyan Dong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| |
Collapse
|
43
|
Choi SY, Yu YS, Park E, Baek SH, Nam JM. Cell-Interface-Deciphering Lipid Nanotablet for Nanoparticle Logic Gate-Based Real-Time Single-Cell Analysis. NANO LETTERS 2025; 25:2725-2731. [PMID: 39811940 DOI: 10.1021/acs.nanolett.4c05747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Analyzing the cell interface is of paramount importance in understanding how cells interact and communicate with other cells, but an advanced analytical platform that can process complex and networked interactions between cell surface ligands and receptors is lacking. Herein, we developed the cell-interface-deciphering lipid nanotablet (CID-LNT) for multiplexed real-time cell analysis. LNT is a nanoparticle-tethered lipid bilayer chip where freely diffusing plasmonic nanoparticles induce scattering signal changes. The CID-LNT transduces cell surface protein information into DNA data, which operate as nanoparticle logic gates. As a proof of concept, we detected and analyzed programmed death-ligand 1 (PD-L1) and associated immune signals (TNF-α, EGF, and IFN-γ). PD-L1 is an immune checkpoint that suppresses T cell activity with inflammatory biomolecules facilitating its expression. The CID-LNT can serve as a dynamic nanoparticle logic board, enabling the logic gate-based analysis of membrane proteins, and can be expanded to immunological synapse analysis, cell interface engineering, and molecular diagnostics.
Collapse
Affiliation(s)
- So Young Choi
- Department of Chemistry, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Young Suk Yu
- School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Eunhye Park
- Department of Chemistry, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Sung Hee Baek
- School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Jwa-Min Nam
- Department of Chemistry, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| |
Collapse
|
44
|
Zhao M, Zhang J, He Y, You C. ASF1B promotes gastric cancer progression by modulating H2AC20 and activating PI3K/AKT and ERK1/2 pathways. Front Pharmacol 2025; 16:1533257. [PMID: 40041497 PMCID: PMC11876136 DOI: 10.3389/fphar.2025.1533257] [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: 11/23/2024] [Accepted: 01/24/2025] [Indexed: 03/06/2025] Open
Abstract
Background Gastric cancer (GC) ranks among the most prevalent malignant neoplasms globally and is associated with a significant mortality rate. Despite the availability of various therapeutic interventions for GC, the overall prognosis for this disease remains unfavorable. This can be attributed to several factors, including delayed diagnosis and the inherent heterogeneity of the tumors. With the continuous enrichment of treatment methods, GC has entered an era of comprehensive treatment oriented toward precision and standardization. Methods Through the application of bioinformatics and assessments of tissue microarrays, this study has selected the histone chaperone Anti-Silencing Function 1B (ASF1B) for detailed analysis, including clinical specimens. We then constructed ASF1B knockout and overexpression cell lines, and conducted biological function tests on this basis, validated at mouse and organoid levels. Additionally, human immunereconstitution was performed in NOD-PrkdcscidIl2rgem1/Smoc (NSG) mice, followed by flow cytometry analysis of mouse blood. Mechanically, protein-protein interaction analyses were conducted utilizing Immunoprecipitation-Mass Spectrometry (IP-MS) and Tandem mass tagging (TMT) methodologies to identify protein clusters. Results The analysis demonstrated that ASF1B is significantly upregulated in GC tissues and correlates with unfavorable prognostic outcomes. Biological function tests provided that ASF1B contributes to tumor cell proliferation, colony formation, invasion and migration, and plays an important role in the progression of GC in vivo. These findings were validated at both the mouse and organoid levels. Additionally, we observed that ASF1B is involved in the tumor microenvironment, where ASF1B knockdown increases CD8+ T cell infiltration, indicating a negative correlation with immune activation. Mechanically, our investigation revealed that ASF1B emerged as a promoter of GC progression by downregulating H2A clustered histone 20 (H2AC20), thereby influencing the activation of the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) and extracellular regulated protein kinases (ERK)1/2 signaling pathways. Conclusion ASF1B, recognized as an oncogene, contributes to the initiation and progression of tumors, positioning it as a prospective target for therapeutic intervention in GC.
Collapse
Affiliation(s)
- Mengyuan Zhao
- Laboratory Medicine Center, The Second Hospital and Clinical Medical School, Lanhzou University, Lanzhou, China
| | - Junchang Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yanjun He
- Laboratory Medicine Center, The Second Hospital and Clinical Medical School, Lanhzou University, Lanzhou, China
| | - Chongge You
- Laboratory Medicine Center, The Second Hospital and Clinical Medical School, Lanhzou University, Lanzhou, China
| |
Collapse
|
45
|
Fu M, Zhou H, Yang J, Cao D, Yuan Z. Infiltration of CD8 + cytotoxic T-cells and expression of PD-1 and PD-L1 in ovarian clear cell carcinoma. Sci Rep 2025; 15:4716. [PMID: 39922892 PMCID: PMC11807128 DOI: 10.1038/s41598-025-89270-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 02/04/2025] [Indexed: 02/10/2025] Open
Abstract
Ovarian clear cell carcinoma (OCCC) is resistant to chemotherapy, with limited treatment options for advanced and recurrent disease. The prevalence of OCCC differs by region. Assessing the expression of programmed cell death ligand 1 (PD-L1), PD-1, and CD8+T cell infiltration in OCCC is crucial, as their correlation with patient survival may provide valuable prognostic insights. We collected data from 36 samples from 18 OCCC patients, including 18 pairs of tumors and adjacent nonneoplastic samples. The optimized multiplex immunofluorescence technique was used to stain paraffin sections for immune factors related to the immune microenvironment of OCCC and clinical prognosis. The expression of PDL1 and PD1 in the tumor cells and tumor stromal cells was not significantly correlated with prognosis. Professional quantitative pathological analysis software was used to count the CD8+ cytotoxic T-cells in tumor regions and adjacent nonneoplastic regions in postoperative specimens. There were more CD8+ cytotoxic T-cells in the adjacent nonneoplastic areas than in the tumor tissue samples (p < 0.001). Further analysis revealed that a difference in cell density between CD8+ non-tumor-infiltrating lymphocytes (NTILs) and CD8+ tumor-infiltrating lymphocytes (TILs) exceeding 70 cells/mm2 was associated with poorer progression-free survival (PFS) (p = 0.042). In adjacent nonneoplastic regions, worse PFS was significantly observed in patients with high CD8+ T-cell expression in both total and stromal cells than those with low expression (p = 0.012 vs p = 0.007). The presence of CD8+ T-cells had significant potential for predicting the prognosis of patients with OCCC, which lays a foundation for the development of biomarkers for immune checkpoint blockade treatment response in OCCC patients.
Collapse
Affiliation(s)
- Mengdi Fu
- Department of Obstetrics and Gynecology, National Clinical Research Centre for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Huimei Zhou
- Department of Obstetrics and Gynecology, National Clinical Research Centre for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China.
| | - Jiaxin Yang
- Department of Obstetrics and Gynecology, National Clinical Research Centre for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China.
| | - Dongyan Cao
- Department of Obstetrics and Gynecology, National Clinical Research Centre for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China.
| | - Zhen Yuan
- Department of Obstetrics and Gynecology, National Clinical Research Centre for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| |
Collapse
|
46
|
Nedeljković M, Vuletić A, Mirjačić Martinović K. Divide and Conquer-Targeted Therapy for Triple-Negative Breast Cancer. Int J Mol Sci 2025; 26:1396. [PMID: 40003864 PMCID: PMC11855393 DOI: 10.3390/ijms26041396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and malignant type of breast cancer with limited treatment options and poor prognosis. One of the most significant impediments in TNBC treatment is the high heterogeneity of this disease, as highlighted by the detection of several molecular subtypes of TNBC. Each subtype is driven by distinct mutations and pathway aberrations, giving rise to specific molecular characteristics closely connected to clinical behavior, outcomes, and drug sensitivity. This review summarizes the knowledge regarding TNBC molecular subtypes and how it can be harnessed to devise tailored treatment strategies instead of blindly using targeted drugs. We provide an overview of novel targeted agents and key insights about new treatment modalities with an emphasis on the androgen receptor signaling pathway, cancer stem cell-associated pathways, phosphatidylinositol 3-kinase (PI3K)/AKT pathway, growth factor signaling, and immunotherapy.
Collapse
Affiliation(s)
- Milica Nedeljković
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (A.V.); (K.M.M.)
| | | | | |
Collapse
|
47
|
Kaewkedsri P, Intarawichian P, Jessadapattarakul S, Kunprom W, Koonmee S, Thanee M, Somintara O, Wongbuddha A, Chadbunchachai P, Nawapun S, Aphivatanasiri C. Programmed Cell Death Ligand 1 (PD-L1) and Major Histocompatibility Complex Class I (MHC Class I) Expression Patterns and Their Pathologic Associations in Triple-Negative Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:123-143. [PMID: 39936074 PMCID: PMC11812676 DOI: 10.2147/bctt.s506833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
Purpose This study aims to investigate the clinicopathological characteristics of triple-negative breast cancer (TNBC) in relation to programmed cell death ligand 1 (PD-L1) and major histocompatibility complex class I (MHC class I) expression, with a focus on their prognostic significance. Patients and Methods A retrospective analysis was conducted on formalin-fixed paraffin-embedded (FFPE) tissue samples from 148 TNBC patients diagnosed between 2008 and 2021. Immunohistochemical analysis evaluated PD-L1 and MHC class I expression. PD-L1 was assessed using Combine Positive Scores (CPS), with the threshold set at CPS ≥ 1 and CPS ≥ 10. MHC class I expression was categorized into low and high levels. Associations between these markers, clinicopathological features, overall survival (OS), and disease-free survival (DFS) were analyzed. PD-L1 expression was also compared between older FFPE blocks (2008-2018) versus newer blocks (2019-2021). Results PD-L1 expression was observed in 29.1% of cases with a Combined Positive Score (CPS) ≥1 and 8.8% of CPS ≥10 cases. MHC class I expression was evenly split between low and high levels. Older FFPE blocks (2008-2018) showed lower PD-L1 expression than newer blocks (2019-2021). There was no significant association between PD-L1 expression and overall survival (OS) or disease-free survival (DFS). However, high MHC class I expression was strongly associated with improved OS (HR = 0.469, 95% CI: 0.282-0.780, p=0.004). Patients with negative PD-L1 and high MHC class I expression had the most favorable prognosis, with significant OS for CPS ≥1 (HR = 0.447, 95% CI: 0.236-0.846, p=0.013) and CPS ≥10 (HR = 0.516, 95% CI: 0.307-0.869, p=0.013). Conclusion These findings support the potential of PD-L1 and MHC class I expression as prognostic markers for TNBC, offering insights to guide treatment decisions and improve patient outcomes.
Collapse
Affiliation(s)
- Ponkrit Kaewkedsri
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | | | - Waritta Kunprom
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Supinda Koonmee
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Malinee Thanee
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Ongart Somintara
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Anongporn Wongbuddha
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Payia Chadbunchachai
- Department of Radiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Supajit Nawapun
- Department of Radiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | |
Collapse
|
48
|
Brauer J, Tumani M, Frey N, Lehmann LH. The cardio-oncologic burden of breast cancer: molecular mechanisms and importance of preclinical models. Basic Res Cardiol 2025; 120:91-112. [PMID: 39621070 PMCID: PMC11790711 DOI: 10.1007/s00395-024-01090-w] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 02/04/2025]
Abstract
Breast cancer, the most prevalent cancer affecting women worldwide, poses a significant cardio-oncological burden. Despite advancements in novel therapeutic strategies, anthracyclines, HER2 antagonists, and radiation remain the cornerstones of oncological treatment. However, each carries a risk of cardiotoxicity, though the molecular mechanisms underlying these adverse effects differ. Common mechanisms include DNA damage response, increased reactive oxygen species, and mitochondrial dysfunction, which are key areas of ongoing research for potential cardioprotective strategies. Since these mechanisms are also essential for effective tumor cytotoxicity, we explore tumor-specific effects, particularly in hereditary breast cancer linked to BRCA1 and BRCA2 mutations. These genetic variants impair DNA repair mechanisms, increase the risk of tumorigenesis and possibly for cardiotoxicity from treatments such as anthracyclines and HER2 antagonists. Novel therapies, including immune checkpoint inhibitors, are used in the clinic for triple-negative breast cancer and improve the oncological outcomes of breast cancer patients. This review discusses the molecular mechanisms underlying BRCA dysfunction and the associated pathological pathways. It gives an overview of preclinical models of breast cancer, such as genetically engineered mouse models, syngeneic murine models, humanized mouse models, and various in vitro and ex vivo systems and models to study cardiovascular side effects of breast cancer therapies. Understanding the underlying mechanism of cardiotoxicity and developing cardioprotective strategies in preclinical models are essential for improving treatment outcomes and reducing long-term cardiovascular risks in breast cancer patients.
Collapse
Affiliation(s)
- J Brauer
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - M Tumani
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - N Frey
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - L H Lehmann
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
49
|
Si H, Fu X, Hao Y, Wang Y, Lin G, Wang D, Xu C, Zhang Y, Song Z. The influence of PD-L1 expression levels on the efficacy of combination therapy in thymic epithelial tumors. Clin Transl Oncol 2025; 27:542-548. [PMID: 39046681 DOI: 10.1007/s12094-024-03618-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: 04/07/2024] [Accepted: 07/11/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND The significant expression of PD-L1 in thymic epithelial tumors (TETs) has been confirmed, and immunotherapy and its combination therapy have been effective in TETs. However, there is no present evidence that the expression levels of PD-L1 affects the efficacy of combination therapy. Our study aimed to shed light on this relationship. METHODS Patients with thymic epithelial tumors (TETs) from multicenter hospitals were retrospectively identified. Objective response rate (ORR), progression-free survival (PFS), overall survival (OS), and immune-related adverse events (irAEs) in 22 patients were included. We divided the patients the 22 patients with PD-L1 test into three levels (high expression, low expression and no expression) and analyzed the relationship between the levels of PD-L1 expression and the efficacy of combination therapy. RESULTS Combination therapy showed an effective benefit in 22 patients with TETs, the median PFS (mPFS) was 16 months (95% CI: 8.5-23.5) and the median OS (mOS) was 38 months (95% CI: 21.5-54.5). Cox-regressive analysis found whether PD-L1 expression affected the PFS of patients (p = 0.017). Among the patients with PD-L1 expression, the levels of expression were correlated with curative effect (Kruskal-Wallis test, PFS: P = 0.012; OS: P = 0.01), and high expression group was along with better efficacy than low expression (Wilcoxon test, P = 0.01). Moreover, in 17 patients treated with immunotherapy combined with chemotherapy, the expression of PD-L1 was also associated with efficacy (Kruskal-Wallis test, p = 0.021). CONCLUSIONS PD-L1 expression affects the PFS of patients. High expression of PD-L1 patients with TETs responded better to combination therapy, which could provide a therapeutic option in clinic. Besides, other targeted treatments should be considered.
Collapse
Affiliation(s)
- Han Si
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Clinical Trial, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No.1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
| | - Xiaoshuang Fu
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Clinical Trial, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No.1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
| | - Yue Hao
- Department of Clinical Trial, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No.1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
| | - Yina Wang
- Department of Oncology, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, China
| | - Gen Lin
- Department of Medical Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Dong Wang
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210000, China
| | - Chunwei Xu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Yongchang Zhang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Zhengbo Song
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China.
- Department of Clinical Trial, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No.1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
50
|
Sanchez JC, Pierpont TM, Argueta-Zamora D, Wilson K, August A, Cerione RA. PTEN loss in glioma cell lines leads to increased extracellular vesicle biogenesis and PD-L1 cargo in a PI3K-dependent manner. J Biol Chem 2025; 301:108143. [PMID: 39732171 PMCID: PMC11791317 DOI: 10.1016/j.jbc.2024.108143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/10/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024] Open
Abstract
Phosphatase and Tensin Homolog (PTEN) is one of the most frequently lost tumor suppressors in cancer and the predominant negative regulator of the PI3K-AKT signaling axis. A growing body of evidence has highlighted the loss of PTEN with immuno-modulatory functions including the upregulation of the programmed death ligand-1 (PD-L1), an altered tumor-derived secretome that drives an immunosuppressive tumor immune microenvironment and resistance to certain immunotherapies. Given their roles in immunosuppression and tumor growth, we examined whether the loss of PTEN would impact the biogenesis, cargo, and function of extracellular vesicles (EVs) in the context of the anti-tumor associated cytokine interferon-γ. Through genetic and pharmacological approaches, we show that total cellular expression of PD-L1 is regulated by JAK/STAT signaling, not PI3K signaling. Instead, we observe that PTEN loss specifically upregulates cell surface levels of PD-L1 and enhances the biogenesis of EVs enriched with PD-L1 in a PI3K-dependent manner. We demonstrate that because of these changes, EVs derived from glioma cells lacking PTEN have a greater ability to suppress T cell receptor signaling. Taken together, these findings provide important new insights into how the loss of PTEN can contribute to an immunosuppressive tumor immune microenvironment, facilitate immune evasion, and highlight a novel role for PI3K signaling in the regulation of EV biogenesis and the cargo they contain.
Collapse
Affiliation(s)
- Julio C Sanchez
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Timothy M Pierpont
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Dariana Argueta-Zamora
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Kristin Wilson
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Richard A Cerione
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, USA.
| |
Collapse
|