1
|
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
|
2
|
Ababneh E, Velez S, Zhao J. Immune evasion and resistance in breast cancer. Am J Cancer Res 2025; 15:1517-1539. [PMID: 40371160 PMCID: PMC12070088 DOI: 10.62347/pngt6996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/18/2024] [Indexed: 05/16/2025] Open
Abstract
Breast cancer (BC) is the most common malignancy in females with an increasing incidence in the last decade. The previously observed decline in BC mortality rates has also slowed down recently with an increase in the incidence of invasive BC. BC has various molecular subtypes. Among these subtypes, triple-negative breast cancer (TNBC) represents the most aggressive BC, with a poor prognosis. Because lack of the hormonal or human epidermal growth factor receptor 2 (HER2) receptors, TNBC is resistant to hormonal and HER2 targeted therapy effective for other BC subtypes. The good news is that TNBC has recently been considered an immunologically 'hot' tumor. Therefore, immunotherapy, particularly immune checkpoint inhibitor therapy, represents a promising therapeutic approach TNBC. However, a considerable percentage of patients with TNBC do not respond well to immunotherapy, indicating that TNBC seems to adopt several mechanisms to evade immune surveillance. Thus, it is crucial to investigate the mechanisms underlying TNBC immune evasion and resistance to immunotherapy. In this review, we examine and discuss the most recently discovered mechanisms for BC, with a particular focus on TNBC, to evade the immune surveillance via kidnapping the immune checkpoints, suppressing the immune responses in tumor microenvironment and inhibiting the tumor antigen presentation. Evaluation of these mechanisms in BC will hopefully guide future immunotherapeutic research and clinical trials.
Collapse
Affiliation(s)
- Ebaa Ababneh
- Burnett School of Biomedical Science, Medical College, University of Central Florida Orlando, FL, USA
| | - Sarah Velez
- Burnett School of Biomedical Science, Medical College, University of Central Florida Orlando, FL, USA
| | - Jihe Zhao
- Burnett School of Biomedical Science, Medical College, University of Central Florida Orlando, FL, USA
| |
Collapse
|
3
|
Lin YZ, Liu CH, Wu WR, Liao TY, Lee CC, Li HW, Chung FC, Shen YC, Zhuo GY, Liu LC, Cheng WC, Wang SC. Memory-promoting function of miR-379-5p attenuates CD8 + T cell exhaustion by targeting immune checkpoints. J Immunother Cancer 2025; 13:e010363. [PMID: 40221151 PMCID: PMC11997822 DOI: 10.1136/jitc-2024-010363] [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: 08/16/2024] [Accepted: 02/16/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are epigenetic regulators of T cell maturation and exhaustion. However, the mechanisms by which miRNAs influence T cell function in tumor environments remain unclear. This study focuses on miR-379-5p, which counteracts T cell exhaustion and enhances antitumor responses. METHODS Native CD8+ T cells were isolated from the blood of healthy donors and subjected to chronic stimulation to induce exhaustion. RNA sequencing and miRNA sequencing were performed to identify differentially expressed miRNAs. These miRNAs underwent bioinformatics analyses, including DESeq enrichment, immune cell infiltration assessment, and patient prognostic outcomes in The Cancer Genome Atlas data sets to assess their potential involvement in T cell exhaustion and antitumor immunity. The biological functions of miRNA on T cell differentiation, cytotoxic killing, and immune checkpoint regulation were investigated using in vitro assays, OT-I B16F10-OVA models, and patient-derived tumor organoids. RESULTS MiR-379-5p is downregulated in exhausted T cells and negatively associated with exhausted tumor-infiltrating lymphocytes in advanced tumors. It correlates positively with better survival outcomes in breast cancer, cervical cancer and melanoma. In CD8+ T cells, miR-379-5p reduces the expression of immune checkpoint proteins T cell immunoglobulin and mucin-domain containing-3 (TIM3) and T cell immunoreceptor with Ig and ITIM domains (TIGIT) by targeting their 3' untranslated region. Overexpression of miR-379-5p in CD8+ T cell promotes differentiation into memory-like T effector cells and enhances cytotoxic killing of cancer cells. The transcription factor nuclear receptor subfamily 4 group A member 1 (NR4A1) with increased expression in exhausted T cells and negatively regulates miR-379, restoring immune checkpoint expression and suppressing cancer-killing ability. In contrast, OT-I T cells expressing ectopic miR-379-5p show increased cytotoxicity against B16F10-OVA tumors in mice. Autologous T cells isolated from patients with breast cancer transduced with miR-379-5p significantly improve killing of tumor organoids derived from the same patients. CONCLUSIONS MiR-379-5p acts as an epigenetic tumor suppressor by enhancing CD8+ T cell effector functions and suppressing T cell exhaustion. MiR-379-5p could represent a novel marker and strategy for cancer immunotherapy, offering promising avenues for enhancing antitumor immune responses.
Collapse
Affiliation(s)
- You-Zhe Lin
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Chia-Hsin Liu
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
| | - Wan-Rong Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Ting-Yi Liao
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chuan-Chun Lee
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Hong-Wei Li
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
| | - Feng-Chi Chung
- Program for Cancer Biology and Drug Discovery, China Medical University, Taichung, Taiwan
| | - Yi-Chun Shen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Guan-Yu Zhuo
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung, Taiwan
| | - Liang-Chih Liu
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Wei-Chung Cheng
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Program for Cancer Biology and Drug Discovery, China Medical University, Taichung, Taiwan
| | - Shao-Chun Wang
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
- Program for Cancer Biology and Drug Discovery, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
4
|
Bian J, Shao R, Li J, Zhu J, Shao A, Liu C, Lu LV, Pan H, Shi Y, Fang N. Mechanism research of non-coding RNA in immune checkpoint inhibitors therapy. Cancer Sci 2024; 115:3520-3531. [PMID: 39136293 PMCID: PMC11531961 DOI: 10.1111/cas.16309] [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: 02/18/2024] [Revised: 07/05/2024] [Accepted: 07/26/2024] [Indexed: 11/05/2024] Open
Abstract
Immune checkpoint inhibitor (ICI) therapies for tumors of different systems have attained significant achievements and have changed the current situation of tumor treatment due to their therapeutic characteristics of high specificity and low side effects. The immune checkpoint Programmed death 1/Programmed cell death-Ligand 1 (PD-1/PD-L1) axis exerts a vital role in the immune escape of tumor cells. As a result, it has become a key target for tumor immunotherapy. Therefore, to perfect research into potential regulatory factors for the PD-1/PD-L1 axis, in order to understand and illustrate tumor ICI therapy mechanisms, is a significant goal. Moreover, ncRNA has been verified to regulate the PD-1/PD-L1 axis in the tumor immune microenvironment to regulate tumor genesis and development. ncRNAs can improve or decrease the efficacy of ICI therapy by modulating PD-L1 expression. This review aimed to investigate the mechanisms of action of ncRNA in regulating the PD-1/PD-L1 axis in ICI therapy, to provide more efficient immunotherapy for tumors of different systems.
Collapse
Affiliation(s)
- Jie Bian
- Department of OncologyThe Affiliated People's Hospital of Jiangsu UniversityZhenjiangChina
| | - Rui Shao
- Department of PathologyThe Affiliated People's Hospital of Jiangsu UniversityZhenjiangChina
| | - Juan Li
- Department of OncologyThe Affiliated People's Hospital of Jiangsu UniversityZhenjiangChina
| | - Jing‐Feng Zhu
- Department of Thoracic and Cardiovascular SurgeryThe Affiliated People's Hospital of Jiangsu UniversityZhenjiangChina
| | - Ai‐Zhong Shao
- Department of Thoracic and Cardiovascular SurgeryThe Affiliated People's Hospital of Jiangsu UniversityZhenjiangChina
| | - Chao Liu
- Department of Thoracic and Cardiovascular SurgeryThe Affiliated People's Hospital of Jiangsu UniversityZhenjiangChina
| | - L. V. Lu
- Department of Thoracic and Cardiovascular SurgeryThe Affiliated People's Hospital of Jiangsu UniversityZhenjiangChina
| | - Hui‐Wen Pan
- Department of Thoracic and Cardiovascular SurgeryThe Affiliated People's Hospital of Jiangsu UniversityZhenjiangChina
| | - Yi‐Jun Shi
- Department of Thoracic and Cardiovascular SurgeryThe Affiliated People's Hospital of Jiangsu UniversityZhenjiangChina
| | - Na Fang
- Department of OncologyThe Affiliated People's Hospital of Jiangsu UniversityZhenjiangChina
| |
Collapse
|
5
|
Zabeti Touchaei A, Vahidi S. MicroRNAs as regulators of immune checkpoints in cancer immunotherapy: targeting PD-1/PD-L1 and CTLA-4 pathways. Cancer Cell Int 2024; 24:102. [PMID: 38462628 PMCID: PMC10926683 DOI: 10.1186/s12935-024-03293-6] [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: 01/11/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024] Open
Abstract
Immunotherapy has revolutionized cancer treatment by harnessing the power of the immune system to eliminate tumors. Immune checkpoint inhibitors (ICIs) block negative regulatory signals that prevent T cells from attacking cancer cells. Two key ICIs target the PD-1/PD-L1 pathway, which includes programmed death-ligand 1 (PD-L1) and its receptor programmed death 1 (PD-1). Another ICI targets cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). While ICIs have demonstrated remarkable efficacy in various malignancies, only a subset of patients respond favorably. MicroRNAs (miRNAs), small non-coding RNAs that regulate gene expression, play a crucial role in modulating immune checkpoints, including PD-1/PD-L1 and CTLA-4. This review summarizes the latest advancements in immunotherapy, highlighting the therapeutic potential of targeting PD-1/PD-L1 and CTLA-4 immune checkpoints and the regulatory role of miRNAs in modulating these pathways. Consequently, understanding the complex interplay between miRNAs and immune checkpoints is essential for developing more effective and personalized immunotherapy strategies for cancer treatment.
Collapse
Affiliation(s)
| | - Sogand Vahidi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
6
|
García-Giménez JL, Saadi W, Ortega AL, Lahoz A, Suay G, Carretero J, Pereda J, Fatmi A, Pallardó FV, Mena-Molla S. miRNAs Related to Immune Checkpoint Inhibitor Response: A Systematic Review. Int J Mol Sci 2024; 25:1737. [PMID: 38339019 PMCID: PMC10855819 DOI: 10.3390/ijms25031737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/19/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
The advent of immune checkpoint inhibitors (ICIs) has represented a breakthrough in the treatment of many cancers, although a high number of patients fail to respond to ICIs, which is partially due to the ability of tumor cells to evade immune system surveillance. Non-coding microRNAs (miRNAs) have been shown to modulate the immune evasion of tumor cells, and there is thus growing interest in elucidating whether these miRNAs could be targetable or proposed as novel biomarkers for prognosis and treatment response to ICIs. We therefore performed an extensive literature analysis to evaluate the clinical utility of miRNAs with a confirmed direct relationship with treatment response to ICIs. As a result of this systematic review, we have stratified the miRNA landscape into (i) miRNAs whose levels directly modulate response to ICIs, (ii) miRNAs whose expression is modulated by ICIs, and (iii) miRNAs that directly elicit toxic effects or participate in immune-related adverse events (irAEs) caused by ICIs.
Collapse
Affiliation(s)
- José Luis García-Giménez
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain; (J.L.G.-G.); (F.V.P.)
- INCLIVA Health Research Institute, INCLIVA, 46010 Valencia, Spain
- Consortium Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, 46010 Valencia, Spain
| | - Wiam Saadi
- Department of Biology, Faculty of Nature, Life and Earth Sciences, University of Djillali Bounaama, Khemis Miliana 44225, Algeria;
| | - Angel L. Ortega
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain; (A.L.O.); (J.C.); (J.P.)
| | - Agustin Lahoz
- Biomarkers and Precision Medicine Unit, Health Research Institute-Hospital La Fe, 46026 Valencia, Spain;
- Analytical Unit, Health Research Institute-Hospital La Fe, 46026 Valencia, Spain
| | - Guillermo Suay
- Medical Oncology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain;
| | - Julián Carretero
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain; (A.L.O.); (J.C.); (J.P.)
| | - Javier Pereda
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain; (A.L.O.); (J.C.); (J.P.)
| | - Ahlam Fatmi
- Department of Microbiology & Biochemistry, Faculty of Science, University of M’sila, M’sila 28000, Algeria;
| | - Federico V. Pallardó
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain; (J.L.G.-G.); (F.V.P.)
- INCLIVA Health Research Institute, INCLIVA, 46010 Valencia, Spain
- Consortium Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, 46010 Valencia, Spain
| | - Salvador Mena-Molla
- INCLIVA Health Research Institute, INCLIVA, 46010 Valencia, Spain
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain; (A.L.O.); (J.C.); (J.P.)
| |
Collapse
|
7
|
Zhao W, Li X, Ren Q, Wang Q, Liao C, Ding T, Li P, Liu J. Molecular mechanism of miRNA regulating PD-L1 expression. GENE REPORTS 2023. [DOI: 10.1016/j.genrep.2023.101763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
8
|
Liu Z, Yu X, Xu L, Li Y, Zeng C. Current insight into the regulation of PD-L1 in cancer. Exp Hematol Oncol 2022; 11:44. [PMID: 35907881 PMCID: PMC9338491 DOI: 10.1186/s40164-022-00297-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/19/2022] [Indexed: 12/09/2023] Open
Abstract
The molecular mechanisms underlying cancer immune escape are a core topic in cancer immunology research. Cancer cells can escape T cell-mediated cellular cytotoxicity by exploiting the inhibitory programmed cell-death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1, CD274) immune checkpoint. Studying the PD-L1 regulatory pattern of tumor cells will help elucidate the molecular mechanisms of tumor immune evasion and improve cancer treatment. Recent studies have found that tumor cells regulate PD-L1 at the transcriptional, post-transcriptional, and post-translational levels and influence the anti-tumor immune response by regulating PD-L1. In this review, we focus on the regulation of PD-L1 in cancer cells and summarize the underlying mechanisms.
Collapse
Affiliation(s)
- Zhuandi Liu
- The First Affiliated Hospital, Institute of Hematology, School of Medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangzhou, China.,Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, 510632, Guangdong, China
| | - Xibao Yu
- The First Affiliated Hospital, Institute of Hematology, School of Medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangzhou, China.,Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, 510632, Guangdong, China
| | - Ling Xu
- The First Affiliated Hospital, Institute of Hematology, School of Medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangzhou, China.,Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, 510632, Guangdong, China
| | - Yangqiu Li
- The First Affiliated Hospital, Institute of Hematology, School of Medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangzhou, China. .,Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, 510632, Guangdong, China.
| | - Chengwu Zeng
- The First Affiliated Hospital, Institute of Hematology, School of Medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangzhou, China. .,Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, 510632, Guangdong, China.
| |
Collapse
|