1
|
Zhu W, Yi Q, Chen Z, Wang J, Zhong K, Ouyang X, Yang K, Jiang B, Zhong J, Zhong J. Exploring the role and mechanisms of MAGEA4 in tumorigenesis, regulation, and immunotherapy. Mol Med 2025; 31:43. [PMID: 39905312 PMCID: PMC11796067 DOI: 10.1186/s10020-025-01079-8] [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: 09/13/2024] [Accepted: 01/13/2025] [Indexed: 02/06/2025] Open
Abstract
MAGEA4 is a member of the Melanoma-Associated Antigen (MAGE) family, characterized by high expression in various tumor tissues but low expression in normal tissues, with the exception of testis and placenta. Its expression is associated with poor prognosis in cancer. This review summarizes the mechanisms of action, regulatory functions, and immunotherapeutic applications of MAGEA4 in cancer.MAGEA4 promotes tumor initiation and progression through multiple pathways, including ubiquitination and degradation of the tumor suppressor P53, regulation of cell cycle and apoptosis, modulation of DNA damage repair, and enhancement of cancer cell survival. By forming a complex with TRIM28, MAGEA4 accelerates tumor development via P53 degradation. Factors such as TWIST1 and BORIS can upregulate MAGEA4 expression. MAGEA4 interacts with proteins including Miz-1, p53, and RAD18, participating in gene transcription regulation and DNA damage repair. By stabilizing RAD18, MAGEA4 facilitates the recruitment of Y-family DNA polymerases, enabling cells to continue replication under DNA damage conditions and thus supporting cancer cell survival. MAGEA4-based TCR-T cell therapy and cancer vaccines show clinical potential. This article comprehensively reviews the structure and function of MAGEA4, as well as recent research progress in solid tumors, providing a theoretical foundation for the clinical translation of MAGEA4 and its application in immunotherapy.
Collapse
Affiliation(s)
- Weijian Zhu
- Gannan Medical University, Ganzhou, Jiangxi, China
| | - Qiang Yi
- Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zheng Chen
- Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jiaqi Wang
- Gannan Medical University, Ganzhou, Jiangxi, China
| | - Kui Zhong
- Gannan Medical University, Ganzhou, Jiangxi, China
| | | | - Kuan Yang
- Gannan Medical University, Ganzhou, Jiangxi, China
| | - Bowei Jiang
- Gannan Medical University, Ganzhou, Jiangxi, China
| | | | - Jinghua Zhong
- Department of Oncology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China.
| |
Collapse
|
2
|
Chandora K, Chandora A, Saeed A, Cavalcante L. Adoptive T Cell Therapy Targeting MAGE-A4. Cancers (Basel) 2025; 17:413. [PMID: 39941782 PMCID: PMC11815873 DOI: 10.3390/cancers17030413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
MAGE A4 (Melanoma Antigen Gene A4) is a cancer testis antigen (CTA) that is expressed normally in germline cells (testis/embryonic tissues) but absent in somatic cells. The MAGE A4 CTA is expressed in a variety of tumor types, like synovial sarcoma, ovarian cancer and non-small cell lung cancer. Having its expression profile limited to germline cells has made MAGE A4 a sought-after immunotherapeutic target in certain malignancies. In this review, we focus on MAGE-A4's function and expression, current clinical trials involving targeted immunotherapy approaches, and challenges and opportunities facing MAGE-A4's targeted therapeutics.
Collapse
Affiliation(s)
- Kapil Chandora
- Morehouse School of Medicine, 720 Westview Dr, Atlanta, GA 30310, USA; (K.C.)
| | - Akshay Chandora
- Morehouse School of Medicine, 720 Westview Dr, Atlanta, GA 30310, USA; (K.C.)
| | - Anwaar Saeed
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA;
| | - Ludimila Cavalcante
- Division of Hematology and Oncology, University of Virginia Comprehensive Cancer Center, Charlottesville, VA 22903, USA
| |
Collapse
|
3
|
Li L, Zhang X, Yan J, Guo J, Liu F, Wei X, Liu Q, Wang K, Liu B. A panel of cancer testis antigens in squamous cell carcinoma of the lung, head and neck, and esophagus: implication for biomarkers and therapeutic targets. Discov Oncol 2025; 16:88. [PMID: 39864021 PMCID: PMC11769918 DOI: 10.1007/s12672-025-01804-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
This study aims to investigate the expression of seven cancer testis antigens (MAGE-A1, MAGE-A4, MAGE-A10, MAGE-A11, PRAME, NY-ESO-1 and KK-LC-1) in pan squamous cell carcinoma and their prognostic value, thus assessing the potential of these CTAs as immunotherapeutic targets. The protein expression of these CTAs was evaluated by immunohistochemistry in 60 lung squamous cell carcinoma (LUSC), 62 esophageal squamous cell carcinoma (ESCA) and 62 head and neck squamous cell carcinoma (HNSC). The relationship between CTAs expression and progression-free survival (PFS) was assessed. PD-L1 expression and tumor-infiltrating lymphocytes were also collected and correlated with CTAs expression. The prognostic impact of CTAs gene expression was evaluated using the Kaplan-Meier Plotter website. CTAs expression was 0-48% in ESCA, 3%-77% in LUSC, and 3%-71% in HNSC. Analysis of PFS showed that MAGE-A1 expression in HNSC (**p < 0.01), PRAME in LUSC (p = 0.008, **p < 0.01), MAGE-A10 (p = 0.012, *p < 0.05) and PRAME (p = 0.021, *p < 0.05) in ESCA were significantly correlated with PFS. In all three cancers, coexpression of three CTAs was used as a cutoff value for grouping, and the results showed a significant difference in PFS between these two groups. Moreover, CTAs expression was significantly correlated with PD-L1 expression and T cell infiltration. These findings indicate a high incidence of CTA expression in HNSC, LUSC and ESCA, which was correlated with PD-L1 expression, T cell infiltration, and tumor progression. The results suggest that cancer testis antigens could be feasible vaccine targets in squamous cell carcinoma.
Collapse
Affiliation(s)
- Lin Li
- Department of Pathology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Xin Zhang
- Department of Pathology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Jiayao Yan
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Jingyi Guo
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of China Pharmaceutical University, Nanjing, 210008, Jiangsu, China
| | - Fangcen Liu
- Department of Pathology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Xiao Wei
- Department of Pathology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Qin Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Kongcheng Wang
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, China.
| | - Baorui Liu
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China.
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, China.
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of China Pharmaceutical University, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
4
|
Cen L, Zhang Z, Sun Y, Wu N, Shao J, Qian Z, Tian M, Ke Y, Liu B. Efficacy of MAGE-A4 long peptide as a universal immunoprevention cancer vaccine. Cancer Cell Int 2024; 24:232. [PMID: 38961429 PMCID: PMC11223347 DOI: 10.1186/s12935-024-03421-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/22/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND The clinical application of peptide vaccines in tumor immunotherapy holds significant promise. Peptide-based tumor vaccines are currently subject to certain limitations in clinical trials, including the challenge of inducing a sustained response from CD4+ T helper cells and cytotoxic T lymphocytes (CTL), as well as human leukocyte antigen (HLA) restrictions. METHODS Through the utilization of biological information methodology, a screening process was conducted to identify three potential long peptides that are specifically targeted by the MAGE-A4 antigen. The candidate long peptides were subjected to in vitro testing using human peripheral blood lymphocytes as samples to evaluate their immunogenicity and immune function. The antitumor properties and preliminary mechanism of the long peptide vaccine were investigated through the use of a mouse model designed for the prevention of triple negative breast cancer (TNBC). RESULTS Three predicted multi-epitope long peptides targeting MAGE-A4 have shown to have a strong immunogenicity, with a total positive rate of 72% across different HLA subtypes in Chinese populations. they can also increase the levels of the costimulatory factor CD137 and tumor necrosis factor-alpha (TNF-α), activate T cells, and boost the cytotoxic activity. Results from an animal study have revealed that the long-peptide vaccine, both on its own and in combination with R848, has displayed impressive anti-tumor and target-specific capabilities. Moreover, it has the ability to increase the expression of effector memory T cells and central memory T cells. CONCLUSIONS This study was the first to screen three multi-epitope long peptides targeting MAGE-A4 and assess their immunogenicity, immune function, and potential as adjuvant peptides. The results showed that the MAGE-A4 long peptide vaccine can be used as a novel immunoprophylaxis method to prevent TNBC. Moreover, the proposed development model is capable of screening multiple target antigens, which lead to its clinical application.
Collapse
Affiliation(s)
- Lanqi Cen
- Department of Oncology, China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, 210000, China
| | - Zhe Zhang
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Yi Sun
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210000, China
| | - Nandie Wu
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210000, China
| | - Jie Shao
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210000, China
| | - Zhaoye Qian
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210000, China
| | - Manman Tian
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210000, China
| | - Yaohua Ke
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210000, China
| | - Baorui Liu
- Department of Oncology, China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, 210000, China.
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210000, China.
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210000, China.
| |
Collapse
|
5
|
Bai R, Yuan C. Kita-Kyushu Lung Cancer Antigen-1 (KK-LC-1): A Promising Cancer Testis Antigen. Aging Dis 2022; 13:1267-1277. [PMID: 35855340 PMCID: PMC9286905 DOI: 10.14336/ad.2021.1207] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/07/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer has always been a huge problem in the field of human health, and its early diagnosis and treatment are the key to solving this problem. Cancer testis antigens (CTAs) are a family of multifunctional proteins that are specifically expressed in male spermatozoa and tumor cells but not in healthy somatic cells. Studies have found that CTAs are involved in the occurrence and development of tumors, and some CTAs trigger immunogenicity, which suggests a possibility of tumor immunotherapy. The differential expression and function of CTAs in normal tissues and tumor cells can promote the screening of tumor markers and the development of new immunotherapies. This article introduces the expression of Kita-Kyushu lung cancer antigen-1 (KK-LC-1), a new member of the CTA family, in different types of tumors and its role in immunotherapy.
Collapse
Affiliation(s)
- Rui Bai
- 1Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Cheng Yuan
- 2Department of Gynecological Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
6
|
Kaneda T, Kurata T, Yoshida T, Kibata K, Yoshioka H, Yanagimoto H, Takeda K, Yoshida T, Tsuta K. Massive digital gene expression analysis reveals different predictive profiles for immune checkpoint inhibitor therapy between adenocarcinoma and squamous cell carcinoma of advanced lung cancer. BMC Cancer 2022; 22:154. [PMID: 35135489 PMCID: PMC8822674 DOI: 10.1186/s12885-022-09264-2] [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: 10/31/2021] [Accepted: 01/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors prolong the survival of non-small cell lung cancer (NSCLC) patients. Although it has been acknowledged that there is some correlation between the efficacy of anti-programmed cell death-1 (PD-1) antibody therapy and immunohistochemical analysis, this technique is not yet considered foolproof for predicting a favorable outcome of PD-1 antibody therapy. We aimed to predict the efficacy of nivolumab based on a comprehensive analysis of RNA expression at the gene level in advanced NSCLC. METHODS This was a retrospective study on patients with NSCLC who were administered nivolumab at the Kansai Medical University Hospital. To identify genes associated with response to anti-PD-1 antibodies, we grouped patients into responders (complete and partial response) and non-responders (stable and progressive disease) to nivolumab therapy. Significant genes were then identified for these groups using Welch's t-test. RESULTS Among 42 analyzed cases (20 adenocarcinomas and 22 squamous cell carcinomas), enhanced expression of MAGE-A4, BBC3, and OTOA genes was observed in responders with adenocarcinoma, and enhanced expression of DAB2, HLA-DPB,1 and CDH2 genes was observed in responders with squamous cell carcinoma. CONCLUSIONS This study predicted the efficacy of nivolumab based on a comprehensive analysis of mRNA expression at the gene level in advanced NSCLC. We also revealed different gene expression patterns as predictors of the effectiveness of anti PD-1 antibody therapy in adenocarcinoma and squamous cell carcinoma.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/immunology
- Adenocarcinoma/drug therapy
- Adenocarcinoma/immunology
- Adult
- Aged
- Aged, 80 and over
- Antigens, CD/immunology
- Antigens, Neoplasm/immunology
- Apoptosis Regulatory Proteins/immunology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/immunology
- Cadherins/immunology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/immunology
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/immunology
- Female
- GPI-Linked Proteins/immunology
- Gene Expression/drug effects
- Gene Expression/immunology
- HLA-DP beta-Chains/immunology
- Humans
- Immune Checkpoint Inhibitors/therapeutic use
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Male
- Middle Aged
- Neoplasm Proteins/immunology
- Nivolumab/therapeutic use
- Predictive Value of Tests
- Programmed Cell Death 1 Receptor/drug effects
- Programmed Cell Death 1 Receptor/immunology
- Proto-Oncogene Proteins/immunology
- RNA, Messenger/drug effects
- RNA, Messenger/immunology
- Retrospective Studies
- Treatment Outcome
Collapse
Affiliation(s)
- Toshihiko Kaneda
- Department of Thoracic Oncology, Kansai Medical University Hospital, 2-3-1, Shinmachi, Hirakata City, Osaka, 573-1191, Japan.
| | - Takayasu Kurata
- Department of Thoracic Oncology, Kansai Medical University Hospital, 2-3-1, Shinmachi, Hirakata City, Osaka, 573-1191, Japan
| | - Tomoko Yoshida
- Discovery Technology Research, Ono Pharmaceutical, Co., Ltd., Osaka, Japan
| | - Kayoko Kibata
- Department of Thoracic Oncology, Kansai Medical University Hospital, 2-3-1, Shinmachi, Hirakata City, Osaka, 573-1191, Japan
| | - Hiroshige Yoshioka
- Department of Thoracic Oncology, Kansai Medical University Hospital, 2-3-1, Shinmachi, Hirakata City, Osaka, 573-1191, Japan
| | - Hiroaki Yanagimoto
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
- Corporate Sponsored Research Programs for Cancer Immunogenomics, Kansai Medical University, Osaka, Japan
| | - Kazuhiko Takeda
- Research Center of Oncology, Ono Pharmaceutical, Co., Ltd., Osaka, Japan
| | - Takao Yoshida
- Research Center of Oncology, Ono Pharmaceutical, Co., Ltd., Osaka, Japan
| | - Koji Tsuta
- Corporate Sponsored Research Programs for Cancer Immunogenomics, Kansai Medical University, Osaka, Japan
- Department of Pathology, Kansai Medical University, Osaka, Japan
| |
Collapse
|
7
|
Herrera LRM. Reverse Vaccinology Approach in Constructing a Multi-Epitope Vaccine Against Cancer-Testis Antigens Expressed in Non-Small Cell Lung Cancer. Asian Pac J Cancer Prev 2021; 22:1495-1506. [PMID: 34048178 PMCID: PMC8408400 DOI: 10.31557/apjcp.2021.22.5.1495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Indexed: 12/26/2022] Open
Abstract
Background: The 5-year survival rate of non-small cell lung cancer (NSCLC) patients has not significantly improved despite advancements in the currently applied treatments. Thus, efforts are put forth in developing novel immunotherapeutic agents targeting cancer-testis antigens (CTA) in NSCLC. This work utilized reverse vaccinology approach in designing a novel multi-epitope vaccine targeting melanoma-associated antigen 3 (MAGEA3), MAGEA4, New York esophageal squamous cell carcinoma-1 (NY-ESO-1), and Kita-Kyushu lung cancer antigen 1 (KK-LC1), being the most frequently expressed CTAs in NSCLC. Methods: Epitopes were mapped from the sequences of CTAs. The population coverage (PC) of identified CD4+ and CD8+ epitopes were estimated. Candidate linear B cell (BL), CD4+, and CD8+ epitopes were adjoined in a multi-epitope construct (Mvax) with flagellin domain as an adjuvant. Antigenicity, and cross-reactivity of Mvax were examined. The tertiary structure of Mvax was modelled, and validated. All epitopes included in the vaccine were docked with their human leukocyte antigen (HLA) binders. The immunogenicity of epitopes in Mvax was validated through molecular dynamics analysis. Results: Mvax contains 22 epitopes from MAGEA3, MAGEA4, NY-ESO-1, and KK-LC1. It is classified as antigenic, non-allergen, non-toxic, and possesses physicochemical stability. Epitopes have no significant hits with other human proteins, except for 2 other CTAs frequently expressed in NSCLC. The stretch of BL epitopes in Mvax confers flexibility, and accessibility emphasizing its antigenicity. The tertiary structure analysis showed that Mvax model has good structural quality. All epitopes included in the vaccine are highly immunogenic as indicated by favorable binding affinity, low binding energy, and acceptable root-mean-square deviation (RMSD). CD4+ and CD8+ epitopes have global PC of 81.81%, and 84.15%, respectively. Conclusion: Overall, in silico evaluations show that Mvax is a potential immunotherapeutic agent against NSCLC.
Collapse
Affiliation(s)
- Leana Rich M Herrera
- Department of Physical Sciences, College of Science, Polytechnic University of the Philippines, Manila City, Philippines
| |
Collapse
|