1
|
Knafler G, Ho AL, Moore KN, Pollack SM, Navenot JM, Sanderson JP. Melanoma-associated antigen A4: A cancer/testis antigen as a target for adoptive T-cell receptor T-cell therapy. Cancer Treat Rev 2025; 134:102891. [PMID: 39970827 DOI: 10.1016/j.ctrv.2025.102891] [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: 11/15/2024] [Revised: 01/24/2025] [Accepted: 01/25/2025] [Indexed: 02/21/2025]
Abstract
T-cell receptor (TCR) T-cell therapies are adoptive cell therapies in which patient cells are engineered to express TCRs targeting specific cancer antigens and infused back into the patient. Since TCR recognition depends on antigen presentation by the human leukocyte antigen system, TCRs can respond to intracellular antigens. Cancer/testis antigens (CTAs) are a large family of proteins, many of which are only expressed in cancerous tissue and immune-privileged germline sites. Melanoma-associated antigen A4 (MAGE-A4) is an intracellular CTA expressed in healthy testis and placenta, and in a range of cancers, including esophageal, head and neck, gastric, ovarian, colorectal, lung, endometrial, cervical, bladder, breast and prostate cancers; soft tissue sarcomas; urothelial and hepatocellular carcinomas; osteosarcoma; and melanoma. This expression pattern, along with the immunogenicity and potential role in tumorigenesis of MAGE-A4 make it a prime target for TCR T-cell therapy. We outline the preclinical and clinical development of TCR T-cell therapies targeting CTAs for treatment of solid tumors, highlighting the need for extensive preclinical characterization of putative off-target, and potential on-target but off-tumor, effects. We identified ten clinical trials assessing TCR T-cell therapies targeting MAGE-A4. Overall, manageable safety profiles and signals of efficacy have been observed, especially in patients with advanced synovial sarcoma, myxoid/round cell liposarcoma, ovarian, head and neck, and urothelial cancers, with one TCR T-cell therapy approved by the US Food and Drug Administration in August 2024. We also review the limitations, and strategies to enhance efficacy and improve safety, of these therapies, and summarize related immunotherapies targeting MAGE-A4.
Collapse
Affiliation(s)
| | - Alan L Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Medical College of Cornell University New York NY USA
| | - Kathleen N Moore
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center Oklahoma City OK USA
| | - Seth M Pollack
- Lurie Cancer Center, Department of Medicine, Northwestern University Feinberg School of Medicine Chicago IL USA
| | | | | |
Collapse
|
2
|
Liu YC, Yu CH, Wang QY, Zhong JT, Bao YY, Fu ZM, Chen Z, Chen HC, Cao ZZ, Zhou SH. MAGEA9 Expression in Vocal Fold Leukoplakia and Its Enhancement of Vocal Fold Leukoplakia Epithelial Cell Proliferation, Migration, and Invasion Through the NF-kB-MMP-2/9 Pathway. J Voice 2025:S0892-1997(25)00042-6. [PMID: 39979188 DOI: 10.1016/j.jvoice.2025.02.001] [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: 12/30/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/22/2025]
Abstract
PURPOSE The expression of MAGE-A9 in vocal fold leukoplakia (VFL) tissues and the mechanism underlying its role in the malignant transformation of VFL remain unclear. This study investigated the role of MAGE-A9 in the proliferation, migration, and invasion of VFL epithelial cells as well as the underlying regulatory mechanism. METHODS MAGE-A9 expression in VFL tissues was detected by immunohistochemistry. The CCK-8 assay, flow cytometry, and transwell assays were performed to determine the viability, cell cycle distribution and apoptosis, as well as migration and invasion, respectively, of primary cultured VFL epithelial cells. Ki67, cell cycle proteins, and proteins in the NF-kB-MMP-2/9 pathway were assessed by Western blotting. RESULTS MAGE-A9 expression was detected in 54.5% (18/33) of vocal fold polyps, 48.9% (44/90) of VFLs, and 84.8% (28/33) of laryngeal cancers. Significantly higher levels of expression were found in laryngeal cancer than in vocal fold polyps and VFL tissues (P < 0.001). The expression of MAGE-A9 tended to increase with the severity of VFL dysplasia. In VFL epithelial cells, the overexpression of MAGE-A9 significantly increased the viability, proliferation, migration, and invasion of the cells and reduced the level of apoptosis. The cell cycle effects of MAGE-A9 overexpression included an increased proportion of cells in the G2 and S phases and a decreased proportion of those in the G1 phase (P = 0.002), leading to an altered G1/S phase transition. MAGE-A9 overexpression also significantly increased p-IKBα, p-p65, MMP2, and MMP9 levels while decreasing those of IKBα. All of the effects of MAGE-A9 were inhibited by treating the cells with the IκBα phosphorylation inhibitor BAY 11-7082. CONCLUSION MAGE-A9 expression tended to increase with the severity of dysplasia in VFL and was significantly higher in laryngeal cancer than in VFL. MAGE-A9 was shown to promote the proliferation, migration, and invasion of VFL epithelial cells via the NF-kB pathway and downstream targets such as MMP-2/9.
Collapse
Affiliation(s)
- Yong-Cai Liu
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, PR China
| | - Chun-Hai Yu
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, PR China
| | - Qin-Ying Wang
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, PR China
| | - Jiang-Tao Zhong
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, PR China
| | - Yang-Yang Bao
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, PR China
| | - Zi-Ming Fu
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, PR China
| | - Zhe Chen
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, PR China
| | - Heng-Chao Chen
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, PR China
| | - Zai-Zai Cao
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, PR China
| | - Shui-Hong Zhou
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, PR China.
| |
Collapse
|
3
|
Buffet A, Filser M, Bruel A, Dard R, Quibel T, Dubucs C, Kwon T, Le Tanno P, Thevenon J, Ziegler A, Allard L, Guigonis V, Roux JJ, Heidet L, Rougeulle C, Boyer O, Vargas-Poussou R, Hureaux M. X-linked transient antenatal Bartter syndrome related to MAGED2 gene: Enriching the phenotypic description and pathophysiologic investigation. Genet Med 2025; 27:101217. [PMID: 39036894 DOI: 10.1016/j.gim.2024.101217] [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: 11/30/2023] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
PURPOSE Transient Bartter syndrome related to pathogenic variants of MAGED2 is the most recently described antenatal Bartter syndrome. Despite its transient nature, it is the most severe form of Bartter syndrome in the perinatal period. Our aim was to describe 14 new cases and to try to explain the incomplete penetrance in women. METHODS We report on 14 new cases, including 3 females, and review the 40 cases described to date. We tested the hypothesis that MAGED2 is transcriptionally regulated by differential methylation of its CpG-rich promotor by pyrosequencing of DNA samples extracted from fetal and adult leukocytes and kidney samples. RESULTS Analysis of the data from 54 symptomatic patients showed spontaneous resolution of symptoms in 27% of cases, persistent complications in 41% of cases, and fatality in 32% of cases. Clinical anomalies were reported in 76% of patients, mostly renal anomalies (52%), cardiovascular anomalies (29%), and dysmorphic features (13%). A developmental delay was reported in 24% of patients. Variants were found in all regions of the gene. Methylation analysis of the MAGED2 CpG-rich promotor showed a correlation with gender, independent of age, tissue or presence of symptoms, excluding a role for this mechanism in the incomplete penetrance in women. CONCLUSION This work enriches the phenotypic and genetic description of this recently described disease and deepens our understanding of the pathophysiological role and regulation of MAGED2. Finally, by describing the wide range of outcomes in patients, this work opens the discussion on genetic counseling offered to families.
Collapse
Affiliation(s)
- Alexandre Buffet
- Département de Médecine Génomique des Tumeurs et Cancers, Hôpital Européen Georges Pompidou, Fédération de Génétique et de Médecine Génomique Assistance Publique-Hôpitaux de Paris Centre Université Paris Cité, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Mathilde Filser
- Département de Médecine Génomique des Tumeurs et Cancers, Hôpital Européen Georges Pompidou, Fédération de Génétique et de Médecine Génomique Assistance Publique-Hôpitaux de Paris Centre Université Paris Cité, Paris, France
| | - Alexandra Bruel
- Service de Pédiatrie, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Rodolphe Dard
- Service de Génétique médicale, Centre Hospitalier Intercommunal de Poissy, Poissy, France
| | - Thibaud Quibel
- Service d'Obstétrique, Centre Hospitalier Intercommunal de Poissy, Poissy, France
| | - Charlotte Dubucs
- Service de Génétique médicale, Oncopole Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Theresa Kwon
- Service de Néphrologie Pédiatrique, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Pauline Le Tanno
- Unité de génétique clinique, Centre Hospitalier Universitaire de Grenoble site Nord - Hôpital Couple-Enfant - Université Grenoble Alpes, La Tranche, France
| | - Julien Thevenon
- Unité de génétique clinique, Centre Hospitalier Universitaire de Grenoble site Nord - Hôpital Couple-Enfant - Université Grenoble Alpes, La Tranche, France
| | - Alban Ziegler
- Service de Génétique Médicale, Centre Hospitalier Universitaire d'Angers, Angers, France; Service de Génétique, CRMR AnDDI-Rares, CHU Reims, Reims, France
| | - Lise Allard
- Service de néphrologie pédiatrique, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Vincent Guigonis
- Service de Néphrologie Pédiatrique, Centre Hospitalier Universitaire de Limoges, Limoges, France
| | - Jean-Jacques Roux
- Laboratoire d'Anatomie et cytologie pathologique, Centre Hospitalier Métropole Savoie, Chambéry, France
| | - Laurence Heidet
- Service de Néphrologie Pédiatrique, Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), Hôpital Necker Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris; Laboratoire des Maladies Rénales Héréditaires, Inserm UMR 1163, Institut Imagine, Université de Paris-Cité, Paris, France
| | - Claire Rougeulle
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, Paris, France
| | - Olivia Boyer
- Université Paris Cité, Inserm, PARCC, Paris, France; Service de Médecine Génomique, Hôpital Européen Georges Pompidou, Fédération de Génétique et de Médecine Génomique Assistance Publique-Hôpitaux de Paris Centre Université Paris Cité, Paris, France
| | - Rosa Vargas-Poussou
- Service de Médecine Génomique, Hôpital Européen Georges Pompidou, Fédération de Génétique et de Médecine Génomique Assistance Publique-Hôpitaux de Paris Centre Université Paris Cité, Paris, France
| | - Marguerite Hureaux
- Université Paris Cité, Inserm, PARCC, Paris, France; Service de Médecine Génomique, Hôpital Européen Georges Pompidou, Fédération de Génétique et de Médecine Génomique Assistance Publique-Hôpitaux de Paris Centre Université Paris Cité, Paris, France; Centre de Recherche Cardio-vasculaire de Paris, UMR970, Paris, France.
| |
Collapse
|
4
|
Mei AHC, Laganà A, Osman R, Cho HJ. Melanoma antigen genes (MAGE); novel functional targets in multiple myeloma. Semin Hematol 2025; 62:43-49. [PMID: 39580273 DOI: 10.1053/j.seminhematol.2024.10.007] [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: 08/30/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 11/25/2024]
Abstract
Melanoma Antigen Genes (MAGE) are expressed in a broad range of cancers, including multiple myeloma. MAGE have been under investigation for more than 3 decades as targets for immune therapy, while in parallel, interrogation of their functions has revealed activities that may be particularly critical in multiple myeloma. MAGE-C1 is expressed in about 75% of newly diagnosed cases and this is maintained through the natural history of the disease. In contrast, MAGE-A3 is expressed in about 35% of newly diagnosed cases, but this increases to more than 75% after relapse. MAGE-A3 expression was associated with poor clinical outcome and resistance to chemotherapy. Translational studies have revealed that MAGE-A3 regulates cell cycling and apoptosis in myeloma cells. Genomic, gene expression, and multiomic studies demonstrate relations with high-risk subgroups of patients. MAGE-A3 mediates these functions through partnership with Kap1 to form a ubiquitin ligase complex. Structural analysis of the interaction between MAGE-A3 and Kap1 gives insight into the biochemical activity and substrate specificity and suggests novel pharmacologic strategies to inhibit them. These studies demonstrating MAGE-A3 oncogenic functions suggest that it may also be a suitable target for small molecule inhibition in multiple myeloma that may be broadly applicable to other cancers that express it.
Collapse
Affiliation(s)
- Anna Huo-Chang Mei
- Multiple Myeloma Center of Excellence, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York NY USA
| | - Alessandro Laganà
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York NY USA
| | - Roman Osman
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Hearn Jay Cho
- Multiple Myeloma Center of Excellence, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York NY USA; The Multiple Myeloma Research Foundation, Norwalk, CT.
| |
Collapse
|
5
|
Zhuo S, Yang S, Chen S, Ding Y, Cheng H, Yang L, Wang K, Yang K. Unveiling the significance of cancer-testis antigens and their implications for immunotherapy in glioma. Discov Oncol 2024; 15:602. [PMID: 39472405 PMCID: PMC11522268 DOI: 10.1007/s12672-024-01449-4] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/11/2024] [Indexed: 11/02/2024] Open
Abstract
Glioma has a poor prognosis, which is attributable to its inherent characteristics and lack of specific treatments. Immunotherapy plays a pivotal role in the contemporary management of malignancies. Despite the initiation of numerous immunotherapy-based clinical trials, their effects on enhancing glioma prognosis remain limited, highlighting the need for innovative and effective therapeutic targets and strategies to address this challenge. Since the 1990s, there has been a growing interest in cancer-testis antigens (CTAs) present in normal mammalian testicular germ cells and placental trophoblast cells, which exhibit reactivated expression in various tumor types. Mechanisms such as DNA methylation, histone modification, transcriptional regulation, and alternative splicing influence the expression of CTAs in tumors. The distinct expression patterns and robust immunogenicity of CTAs are promising tumor biomarkers and optimal targets for immunotherapy. Previous reports have shown that multiple CTAs are present in gliomas and are closely related to prognosis. The expression of these antigens is also associated with the immune response in gliomas and the effectiveness of immunotherapy. Significantly, numerous clinical trials, with IL13RA2 as a representative CTA member, have assessed the immunotherapeutic potential of gliomas and have shown favorable clinical efficacy. This review provides a comprehensive overview of the regulation and function of CTAs, summarizes their expression and role in gliomas, emphasizes their importance as immunotherapy targets in gliomas, and discusses related challenges and future interventions.
Collapse
Affiliation(s)
- Shenghua Zhuo
- Department of Neurosurgery, the First Affiliated Hospital of Hainan Medical University (Hainan Academy of Medical Sciences), Haikou, China.
- International Center for Aging and Cancer, Hainan Medical University (Hainan Academy of Medical Sciences), Haikou, China.
| | - Shuo Yang
- International Center for Aging and Cancer, Hainan Medical University (Hainan Academy of Medical Sciences), Haikou, China
| | - Shenbo Chen
- Department of Neurosurgery, the First Affiliated Hospital of Hainan Medical University (Hainan Academy of Medical Sciences), Haikou, China
| | - Yueju Ding
- Department of Neurosurgery, the First Affiliated Hospital of Hainan Medical University (Hainan Academy of Medical Sciences), Haikou, China
| | - Honglei Cheng
- Department of Neurosurgery, the First Affiliated Hospital of Hainan Medical University (Hainan Academy of Medical Sciences), Haikou, China
| | - Liangwang Yang
- Department of Neurosurgery, the First Affiliated Hospital of Hainan Medical University (Hainan Academy of Medical Sciences), Haikou, China
| | - Kai Wang
- International Center for Aging and Cancer, Hainan Medical University (Hainan Academy of Medical Sciences), Haikou, China.
| | - Kun Yang
- Department of Neurosurgery, the First Affiliated Hospital of Hainan Medical University (Hainan Academy of Medical Sciences), Haikou, China.
| |
Collapse
|
6
|
Li K, Krone MW, Butrin A, Bond MJ, Linhares BM, Crews CM. Development of Ligands and Degraders Targeting MAGE-A3. J Am Chem Soc 2024; 146:24884-24891. [PMID: 39190582 DOI: 10.1021/jacs.4c05393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Type I melanoma antigen (MAGE) family members are detected in numerous tumor types, and expression is correlated with poor prognosis, high tumor grade, and increased metastasis. Type I MAGE proteins are typically restricted to reproductive tissues, but expression can recur during tumorigenesis. Several biochemical functions have been elucidated for them, and notably, MAGEs regulate proteostasis by serving as substrate recognition modules for E3 ligase complexes. The repertoire of E3 ligase complexes that can be hijacked for targeted protein degradation continues to expand, and MAGE-E3 complexes are an especially attractive platform given their cancer-selective expression. Additionally, type I MAGE-derived peptides are presented on cancer cell surfaces, so targeted MAGE degradation may increase antigen presentation and improve immunotherapy outcomes. Motivated by these applications, we developed novel, small-molecule ligands for MAGE-A3, a type I MAGE that is widely expressed in tumors and associates with TRIM28, a RING E3 ligase. Chemical matter was identified through DNA-encoded library (DEL) screening, and hit compounds were validated for in vitro binding to MAGE-A3. We obtained a cocrystal structure with a DEL analog and hypothesize that the small molecule binds at a dimer interface. We utilized this ligand to develop PROTAC molecules that induce MAGE-A3 degradation through VHL recruitment and inhibit the proliferation of MAGE-A3 positive cell lines. These ligands and degraders may serve as valuable probes for investigating MAGE-A3 biology and as foundations for the ongoing development of tumor-specific PROTACs.
Collapse
Affiliation(s)
- Ke Li
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
| | - Mackenzie W Krone
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
| | - Arseniy Butrin
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
| | - Michael J Bond
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
| | - Brian M Linhares
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
| | - Craig M Crews
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
- Department of Chemistry, Yale University, New Haven, Connecticut 06511, United States
- Department of Pharmacology, Yale University, New Haven, Connecticut 06511, United States
| |
Collapse
|
7
|
Kwok DW, Okada H, Costello JF. Activating the dark genome to illuminate cancer vaccine targets. Nat Genet 2024; 56:1770-1771. [PMID: 39223317 PMCID: PMC11456370 DOI: 10.1038/s41588-024-01850-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Epigenetic therapy awakens myriad transposable elements to generate new antigens that could prime tumor cells for immunotherapy. A new study of glioblastoma discovers indiscriminate awakening in normal cells also and then presents a more selective strategy for potential therapeutic targeting.
Collapse
Affiliation(s)
- Darwin W Kwok
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| | - Joseph F Costello
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
8
|
Kalvapudi S, Pachimatla AG, Seager RJ, Conroy J, Pabla S, Mukherjee S. Cancer/testis antigen expression and co-expression patterns in gastroesophageal adenocarcinoma. Med Oncol 2024; 41:227. [PMID: 39143271 PMCID: PMC11324668 DOI: 10.1007/s12032-024-02475-6] [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: 05/30/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Gastroesophageal adenocarcinoma (GEAC) poses a significant challenge due to its poor prognosis and limited treatment options. Recently, Cancer/testis antigens (CTAs) have emerged as potential therapy targets due to their high expression in tumor cells and their immunogenic nature. We aimed to explore the expression and co-expression of CTAs in GEAC. We analyzed 63 GEAC patients initially and validated our findings in 329 patients from The Cancer Genome Atlas (TCGA) database. CTA expression was measured after RNA sequencing, while clinical information, including survival outcomes and treatment details, was collected from an institutional database. Co-expression patterns among CTAs were determined using Spearman correlation analysis. The majority of the study cohort were male (87%), Caucasian (94%), and had stage IV disease (64%). CTAs were highly prevalent, ranging from 58 to 19%. The MAGE gene family showed the highest expression, consistent across both cohorts. The correlation matrix revealed a distinct cluster of significantly co-expressed genes, including MAGEA3, NY-ESO-1, and others (0.27 ≤ r ≤ 0.73). Survival analysis revealed that individual CTAs were associated with poorer survival outcomes in patients not receiving immunotherapy while showing potential for improved survival in those undergoing immunotherapy, although these findings lacked robust reliability. Our study provides a comprehensive characterization of CTA expression and co-expression in GEAC. The strong correlation among CTAs like MAGE, NY-ESO-1, and GAGE suggests a potential for therapies targeting multiple CTAs simultaneously. Further research, including prospective trials, is warranted to assess the prognostic value of CTAs and their suitability as therapeutic targets.
Collapse
Affiliation(s)
- Sukumar Kalvapudi
- Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14206, USA
| | - Akhil Goud Pachimatla
- Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14206, USA
| | - R J Seager
- Labcorp Oncology, 700 Ellicott Street, Buffalo, NY, 14203, USA
| | - Jeffrey Conroy
- Labcorp Oncology, 700 Ellicott Street, Buffalo, NY, 14203, USA
| | - Sarabjot Pabla
- Labcorp Oncology, 700 Ellicott Street, Buffalo, NY, 14203, USA
| | - Sarbajit Mukherjee
- Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14206, USA.
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
9
|
Camarena ME, Theunissen P, Ruiz M, Ruiz-Orera J, Calvo-Serra B, Castelo R, Castro C, Sarobe P, Fortes P, Perera-Bel J, Albà MM. Microproteins encoded by noncanonical ORFs are a major source of tumor-specific antigens in a liver cancer patient meta-cohort. SCIENCE ADVANCES 2024; 10:eadn3628. [PMID: 38985879 PMCID: PMC11235171 DOI: 10.1126/sciadv.adn3628] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 06/04/2024] [Indexed: 07/12/2024]
Abstract
The expression of tumor-specific antigens during cancer progression can trigger an immune response against the tumor. Here, we investigate if microproteins encoded by noncanonical open reading frames (ncORFs) are a relevant source of tumor-specific antigens. We analyze RNA sequencing data from 117 hepatocellular carcinoma (HCC) tumors and matched healthy tissue together with ribosome profiling and immunopeptidomics data. Combining human leukocyte antigen-epitope binding predictions and experimental validation experiments, we conclude that around 40% of the tumor-specific antigens in HCC are likely to be derived from ncORFs, including two peptides that can trigger an immune response in humanized mice. We identify a subset of 33 tumor-specific long noncoding RNAs expressing novel cancer antigens shared by more than 10% of the HCC samples analyzed, which, when combined, cover a large proportion of the patients. The results of the study open avenues for extending the range of anticancer vaccines.
Collapse
Affiliation(s)
| | - Patrick Theunissen
- Center for Applied Medical Research (CIMA), University of Navarra (UNAV), Pamplona, Spain
| | - Marta Ruiz
- Center for Applied Medical Research (CIMA), University of Navarra (UNAV), Pamplona, Spain
| | - Jorge Ruiz-Orera
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Beatriz Calvo-Serra
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Robert Castelo
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Carla Castro
- Center for Applied Medical Research (CIMA), University of Navarra (UNAV), Pamplona, Spain
| | - Pablo Sarobe
- Center for Applied Medical Research (CIMA), University of Navarra (UNAV), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Cancer Clinic University of Navarra (CCUN), Pamplona, Spain
| | - Puri Fortes
- Center for Applied Medical Research (CIMA), University of Navarra (UNAV), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Cancer Clinic University of Navarra (CCUN), Pamplona, Spain
- Spanish Network for Advanced Therapies (TERAV ISCIII), Madrid, Spain
| | | | - M Mar Albà
- Hospital del Mar Research Institute, Barcelona, Spain
- Catalan Institute for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
10
|
Lin L, Zou X, Nong W, Ge Y, Li F, Luo B, Zhang Q, Xie X. The potential value of cancer-testis antigens in ovarian cancer: Prognostic markers and targets for immunotherapy. Immun Inflamm Dis 2024; 12:e1284. [PMID: 38896069 PMCID: PMC11186301 DOI: 10.1002/iid3.1284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/11/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Tumor immunotherapy has become an important adjuvant therapy after surgery, radiotherapy, and chemotherapy. In recent years, the role of tumor-associated antigen (TAA) in tumor immunotherapy has become increasingly prominent. Cancer-testis antigen (CTA) is a kind of TAA that is highly restricted in a variety of tumors and can induce an immune response. AIMS This review article aimed to evaluate the role of CTA on the progression of ovarian cancer, its diagnostic efficacy, and the potential for immunotherapy. METHODS We analyzed publications and outlined a comprehensive of overview the regulatory mechanism, immunogenicity, clinical expression significance, tumorigenesis, and application prospects of CTA in ovarian cancer, with a particular focus on recent progress in CTA-based immunotherapy. RESULTS The expression of CTA affects the occurrence, development, and prognosis of ovarian cancer and is closely related to tumor immunity. CONCLUSION CTA can be used as a biomarker for the diagnosis and prognosis evaluation of ovarian cancer and is an ideal target for antitumor immunotherapy. These findings provide novel insights on CTA in the improvement of diagnosis and treatment for ovarian cancer. The successes, current challenges and future prospects were also discussed to portray its significant potential.
Collapse
Affiliation(s)
- Lina Lin
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Xiaoqiong Zou
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Weixia Nong
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Yingying Ge
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Feng Li
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Bin Luo
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
- Education Department of Guangxi Zhuang Autonomous RegionKey Laboratory of Basic Research on Regional Diseases (Guangxi Medical University)NanningGuangxiPeople's Republic of China
| | - Qingmei Zhang
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
- Education Department of Guangxi Zhuang Autonomous RegionKey Laboratory of Basic Research on Regional Diseases (Guangxi Medical University)NanningGuangxiPeople's Republic of China
| | - Xiaoxun Xie
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
- Education Department of Guangxi Zhuang Autonomous RegionKey Laboratory of Basic Research on Regional Diseases (Guangxi Medical University)NanningGuangxiPeople's Republic of China
- Ministry of Education, Key Laboratory of Early Prevention and Treatment of Regional High Frequency Tumor (Guangxi Medical University)NanningGuangxiPeople's Republic of China
| |
Collapse
|
11
|
Klebanoff CA, Chandran SS, Baker BM, Quezada SA, Ribas A. T cell receptor therapeutics: immunological targeting of the intracellular cancer proteome. Nat Rev Drug Discov 2023; 22:996-1017. [PMID: 37891435 PMCID: PMC10947610 DOI: 10.1038/s41573-023-00809-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2023] [Indexed: 10/29/2023]
Abstract
The T cell receptor (TCR) complex is a naturally occurring antigen sensor that detects, amplifies and coordinates cellular immune responses to epitopes derived from cell surface and intracellular proteins. Thus, TCRs enable the targeting of proteins selectively expressed by cancer cells, including neoantigens, cancer germline antigens and viral oncoproteins. As such, TCRs have provided the basis for an emerging class of oncology therapeutics. Herein, we review the current cancer treatment landscape using TCRs and TCR-like molecules. This includes adoptive cell transfer of T cells expressing endogenous or engineered TCRs, TCR bispecific engagers and antibodies specific for human leukocyte antigen (HLA)-bound peptides (TCR mimics). We discuss the unique complexities associated with the clinical development of these therapeutics, such as HLA restriction, TCR retrieval, potency assessment and the potential for cross-reactivity. In addition, we highlight emerging clinical data that establish the antitumour potential of TCR-based therapies, including tumour-infiltrating lymphocytes, for the treatment of diverse human malignancies. Finally, we explore the future of TCR therapeutics, including emerging genome editing methods to safely enhance potency and strategies to streamline patient identification.
Collapse
Affiliation(s)
- Christopher A Klebanoff
- Memorial Sloan Kettering Cancer Center (MSKCC), Human Oncology and Pathogenesis Program, New York, NY, USA.
| | - Smita S Chandran
- Memorial Sloan Kettering Cancer Center (MSKCC), Human Oncology and Pathogenesis Program, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, New York, NY, USA
- Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Brian M Baker
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, ID, USA
- The Harper Cancer Research Institute, University of Notre Dame, Notre Dame, ID, USA
| | - Sergio A Quezada
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Achilles Therapeutics, London, UK
| | - Antoni Ribas
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| |
Collapse
|
12
|
Samel A, Väärtnõu F, Verk L, Kurg K, Mutso M, Kurg R. How the Intrinsically Disordered N-Terminus of Cancer/Testis Antigen MAGEA10 Is Responsible for Its Expression, Nuclear Localisation and Aberrant Migration. Biomolecules 2023; 13:1704. [PMID: 38136576 PMCID: PMC10741916 DOI: 10.3390/biom13121704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Melanoma-associated antigen A (MAGEA) subfamily proteins are normally expressed in testis and/or placenta. However, aberrant expression is detected in the tumour cells of multiple types of human cancer. MAGEA expression is mainly observed in cancers that have acquired malignant phenotypes, invasiveness and metastasis, and the expression of MAGEA family proteins has been linked to poor prognosis in cancer patients. All MAGE proteins share the common MAGE homology domain (MHD) which encompasses up to 70% of the protein; however, the areas flanking the MHD region vary between family members and are poorly conserved. To investigate the molecular basis of MAGEA10 expression and anomalous mobility in gel, deletion and point-mutation, analyses of the MAGEA10 protein were performed. Our data show that the intrinsically disordered N-terminal domain and, specifically, the first seven amino acids containing a unique linear motif, PRAPKR, are responsible for its expression, aberrant migration in SDS-PAGE and nuclear localisation. The aberrant migration in gel and nuclear localisation are not related to each other. Hiding the N-terminus with an epitope tag strongly affected its mobility in gel and expression in cells. Our results suggest that the intrinsically disordered domains flanking the MHD determine the unique properties of individual MAGEA proteins.
Collapse
Affiliation(s)
| | | | | | | | | | - Reet Kurg
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia; (A.S.); (F.V.); (L.V.); (K.K.); (M.M.)
| |
Collapse
|
13
|
Ghorani E, Swanton C, Quezada SA. Cancer cell-intrinsic mechanisms driving acquired immune tolerance. Immunity 2023; 56:2270-2295. [PMID: 37820584 DOI: 10.1016/j.immuni.2023.09.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023]
Abstract
Immune evasion is a hallmark of cancer, enabling tumors to survive contact with the host immune system and evade the cycle of immune recognition and destruction. Here, we review the current understanding of the cancer cell-intrinsic factors driving immune evasion. We focus on T cells as key effectors of anti-cancer immunity and argue that cancer cells evade immune destruction by gaining control over pathways that usually serve to maintain physiological tolerance to self. Using this framework, we place recent mechanistic advances in the understanding of cancer immune evasion into broad categories of control over T cell localization, antigen recognition, and acquisition of optimal effector function. We discuss the redundancy in the pathways involved and identify knowledge gaps that must be overcome to better target immune evasion, including the need for better, routinely available tools that incorporate the growing understanding of evasion mechanisms to stratify patients for therapy and trials.
Collapse
Affiliation(s)
- Ehsan Ghorani
- Cancer Immunology and Immunotherapy Unit, Department of Surgery and Cancer, Imperial College London, London, UK; Department of Medical Oncology, Imperial College London Hospitals, London, UK.
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Department of Oncology, University College London Hospitals, London, UK
| | - Sergio A Quezada
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Cancer Immunology Unit, Research Department of Hematology, University College London Cancer Institute, London, UK.
| |
Collapse
|
14
|
Zheng J, Wang H. Highly Efficient Gel Electrophoresis for Accurate Quantification of Nucleic Acid Modifications via in-Gel Digestion with UHPLC-MS/MS. Anal Chem 2023; 95:13407-13411. [PMID: 37642231 DOI: 10.1021/acs.analchem.3c02418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Gel electrophoresis is a powerful technique for the characterization of sequences, sizes and conformations of nucleic acids due to its remarkable separation efficiency. In parallel, liquid chromatography-mass spectrometry (LC-MS) has established itself as a staple tool for the meticulous characterization and accurate quantification of a multitude of DNA modifications. In this study, we devised an in-gel digestion method for coupling gel electrophoresis with LC-MS/MS. This process involves the enzymatic digestion of DNA within the gel by nucleases and release single nucleosides, which subsequently serve as a preprocessing step for (LC-MS/MS) analysis. We demonstrated that ethylenediaminetetraacetic acid (EDTA) in the routine gel electrophoresis buffer reduced the enzymatic digestion efficiency, while Mg2+ could mitigate this inhibition. We further showed EDTA-free gel electrophoresis and the process of digestion of genomic DNA and plasmid DNA within a gel was fluorescently imaged, proving the efficient digestion of DNA. By this improvement, the efficiency of an in-gel digestion could reach 60% or more of the control, compared with direct in-solution digestion. The measured abundances of DNA modifications (5-methylcytosine and N6-methyladenine) via in-gel digestion are consistent with that measured by in-solution digestion. Collectively, we showed an in-gel digestion method, which is a very useful pretreatment technique for the precise quantification of epigenetic modifications in diverse DNA molecules.
Collapse
Affiliation(s)
- Jing Zheng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hailin Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
15
|
Norollahi SE, Vahidi S, Shams S, Keymoradzdeh A, Soleymanpour A, Solymanmanesh N, Mirzajani E, Jamkhaneh VB, Samadani AA. Analytical and therapeutic profiles of DNA methylation alterations in cancer; an overview of changes in chromatin arrangement and alterations in histone surfaces. Horm Mol Biol Clin Investig 2023; 44:337-356. [PMID: 36799246 DOI: 10.1515/hmbci-2022-0043] [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: 05/07/2022] [Accepted: 01/24/2023] [Indexed: 02/18/2023]
Abstract
DNA methylation is the most important epigenetic element that activates the inhibition of gene transcription and is included in the pathogenesis of all types of malignancies. Remarkably, the effectors of DNA methylation are DNMTs (DNA methyltransferases) that catalyze de novo or keep methylation of hemimethylated DNA after the DNA replication process. DNA methylation structures in cancer are altered, with three procedures by which DNA methylation helps cancer development which are including direct mutagenesis, hypomethylation of the cancer genome, and also focal hypermethylation of the promoters of TSGs (tumor suppressor genes). Conspicuously, DNA methylation, nucleosome remodeling, RNA-mediated targeting, and histone modification balance modulate many biological activities that are essential and indispensable to the genesis of cancer and also can impact many epigenetic changes including DNA methylation and histone modifications as well as adjusting of non-coding miRNAs expression in prevention and treatment of many cancers. Epigenetics points to heritable modifications in gene expression that do not comprise alterations in the DNA sequence. The nucleosome is the basic unit of chromatin, consisting of 147 base pairs (bp) of DNA bound around a histone octamer comprised of one H3/H4 tetramer and two H2A/H2B dimers. DNA methylation is preferentially distributed over nucleosome regions and is less increased over flanking nucleosome-depleted DNA, implying a connection between nucleosome positioning and DNA methylation. In carcinogenesis, aberrations in the epigenome may also include in the progression of drug resistance. In this report, we report the rudimentary notes behind these epigenetic signaling pathways and emphasize the proofs recommending that their misregulation can conclude in cancer. These findings in conjunction with the promising preclinical and clinical consequences observed with epigenetic drugs against chromatin regulators, confirm the important role of epigenetics in cancer therapy.
Collapse
Affiliation(s)
- Seyedeh Elham Norollahi
- Cancer Research Center and Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Sogand Vahidi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shima Shams
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Arman Keymoradzdeh
- Department of Neurosurgery, School of Medicine, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Armin Soleymanpour
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Nazanin Solymanmanesh
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ebrahim Mirzajani
- Department of Biochemistry and Biophysics, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Vida Baloui Jamkhaneh
- Department of Veterinary Medicine, Islamic Azad University of Babol Branch, Babol, Iran
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
16
|
Bassal MA. The Interplay between Dysregulated Metabolism and Epigenetics in Cancer. Biomolecules 2023; 13:944. [PMID: 37371524 DOI: 10.3390/biom13060944] [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: 04/24/2023] [Revised: 05/21/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Cellular metabolism (or energetics) and epigenetics are tightly coupled cellular processes. It is arguable that of all the described cancer hallmarks, dysregulated cellular energetics and epigenetics are the most tightly coregulated. Cellular metabolic states regulate and drive epigenetic changes while also being capable of influencing, if not driving, epigenetic reprogramming. Conversely, epigenetic changes can drive altered and compensatory metabolic states. Cancer cells meticulously modify and control each of these two linked cellular processes in order to maintain their tumorigenic potential and capacity. This review aims to explore the interplay between these two processes and discuss how each affects the other, driving and enhancing tumorigenic states in certain contexts.
Collapse
Affiliation(s)
- Mahmoud Adel Bassal
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
17
|
Nin DS, Deng LW. Biology of Cancer-Testis Antigens and Their Therapeutic Implications in Cancer. Cells 2023; 12:cells12060926. [PMID: 36980267 PMCID: PMC10047177 DOI: 10.3390/cells12060926] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Tumour-specific antigens have been an area of interest in cancer therapy since their discovery in the middle of the 20th century. In the era of immune-based cancer therapeutics, redirecting our immune cells to target these tumour-specific antigens has become even more relevant. Cancer-testis antigens (CTAs) are a class of antigens with an expression specific to the testis and cancer cells. CTAs have also been demonstrated to be expressed in a wide variety of cancers. Due to their frequency and specificity of expression in a multitude of cancers, CTAs have been particularly attractive as cancer-specific therapeutic targets. There is now a rapid expansion of CTAs being identified and many studies have been conducted to correlate CTA expression with cancer and therapy-resistant phenotypes. Furthermore, there is an increasing number of clinical trials involving using some of these CTAs as molecular targets in pharmacological and immune-targeted therapeutics for various cancers. This review will summarise the current knowledge of the biology of known CTAs in tumorigenesis and the regulation of CTA genes. CTAs as molecular targets and the therapeutic implications of these CTA-targeted anticancer strategies will also be discussed.
Collapse
Affiliation(s)
- Dawn Sijin Nin
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, MD 7, 8 Medical Drive, Singapore 117596, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
| | - Lih-Wen Deng
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, MD 7, 8 Medical Drive, Singapore 117596, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
- National University Cancer Institute, National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| |
Collapse
|
18
|
The Melanoma-Associated Antigen Family A (MAGE-A): A Promising Target for Cancer Immunotherapy? Cancers (Basel) 2023; 15:cancers15061779. [PMID: 36980665 PMCID: PMC10046478 DOI: 10.3390/cancers15061779] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023] Open
Abstract
Early efforts to identify tumor-associated antigens over the last decade have provided unique cancer epitopes for targeted cancer therapy. MAGE-A proteins are a subclass of cancer/testis (CT) antigens that are presented on the cell surface by MHC class I molecules as an immune-privileged site. This is due to their restricted expression to germline cells and a wide range of cancers, where they are associated with resistance to chemotherapy, metastasis, and cancer cells with an increasing potential for survival. This makes them an appealing candidate target for designing an effective and specific immunotherapy, thereby suggesting that targeting oncogenic MAGE-As with cancer vaccination, adoptive T-cell transfer, or a combination of therapies would be promising. In this review, we summarize and discuss previous and ongoing (pre-)clinical studies that target these antigens, while bearing in mind the benefits and drawbacks of various therapeutic strategies, in order to speculate on future directions for MAGE-A-specific immunotherapies.
Collapse
|
19
|
Kumar A, Emdad L, Fisher PB, Das SK. Targeting epigenetic regulation for cancer therapy using small molecule inhibitors. Adv Cancer Res 2023; 158:73-161. [PMID: 36990539 DOI: 10.1016/bs.acr.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Cancer cells display pervasive changes in DNA methylation, disrupted patterns of histone posttranslational modification, chromatin composition or organization and regulatory element activities that alter normal programs of gene expression. It is becoming increasingly clear that disturbances in the epigenome are hallmarks of cancer, which are targetable and represent attractive starting points for drug creation. Remarkable progress has been made in the past decades in discovering and developing epigenetic-based small molecule inhibitors. Recently, epigenetic-targeted agents in hematologic malignancies and solid tumors have been identified and these agents are either in current clinical trials or approved for treatment. However, epigenetic drug applications face many challenges, including low selectivity, poor bioavailability, instability and acquired drug resistance. New multidisciplinary approaches are being designed to overcome these limitations, e.g., applications of machine learning, drug repurposing, high throughput virtual screening technologies, to identify selective compounds with improved stability and better bioavailability. We provide an overview of the key proteins that mediate epigenetic regulation that encompass histone and DNA modifications and discuss effector proteins that affect the organization of chromatin structure and function as well as presently available inhibitors as potential drugs. Current anticancer small-molecule inhibitors targeting epigenetic modified enzymes that have been approved by therapeutic regulatory authorities across the world are highlighted. Many of these are in different stages of clinical evaluation. We also assess emerging strategies for combinatorial approaches of epigenetic drugs with immunotherapy, standard chemotherapy or other classes of agents and advances in the design of novel epigenetic therapies.
Collapse
|
20
|
Pascucci FA, Escalada MC, Suberbordes M, Vidal C, Ladelfa MF, Monte M. MAGE-I proteins and cancer-pathways: A bidirectional relationship. Biochimie 2022; 208:31-37. [PMID: 36403755 DOI: 10.1016/j.biochi.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/31/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022]
Abstract
Data emerged from the last 20 years of basic research on tumor antigens positioned the type I MAGE (Melanoma Antigen GEnes - I or MAGE-I) family as cancer driver factors. MAGE-I gene expression is mainly restricted to normal reproductive tissues. However, abnormal re-expression in cancer unbalances the cell status towards enhanced oncogenic activity or reduced tumor suppression. Anomalous MAGE-I gene re-expression in cancer is attributed to altered epigenetic-mediated chromatin silencing. Still, emerging data indicate that MAGE-I can be regulated at protein level. Results from different laboratories suggest that after its anomalous re-expression, specific MAGE-I proteins can be regulated by well-known signaling pathways or key cellular processes that finally potentiate the cancer cell phenotype. Thus, MAGE-I proteins both regulate and are regulated by cancer-related pathways. Here, we present an updated review highlighting the recent findings on the regulation of MAGE-I by oncogenic pathways and the potential consequences in the tumor cell behavior.
Collapse
Affiliation(s)
- Franco Andrés Pascucci
- Laboratorio de Oncología Molecular, Departamento de Química Biológica and IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Micaela Carolina Escalada
- Laboratorio de Oncología Molecular, Departamento de Química Biológica and IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Melisa Suberbordes
- Laboratorio de Oncología Molecular, Departamento de Química Biológica and IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Candela Vidal
- Laboratorio de Oncología Molecular, Departamento de Química Biológica and IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Fátima Ladelfa
- Laboratorio de Oncología Molecular, Departamento de Química Biológica and IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Martín Monte
- Laboratorio de Oncología Molecular, Departamento de Química Biológica and IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
21
|
Zheng X, Zhao X. A hypothetical model of skewed DNA methylation balance in the enhancer regions containing differentially methylated cytosines associated with non-malignant complex diseases. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
22
|
Tsai JW, Cejas P, Wang DK, Patel S, Wu DW, Arounleut P, Wei X, Zhou N, Syamala S, Dubois FP, Crane A, Pelton K, Vogelzang J, Sousa C, Baguette A, Chen X, Condurat AL, Dixon-Clarke SE, Zhou KN, Lu SD, Gonzalez EM, Chacon MS, Digiacomo JJ, Kumbhani R, Novikov D, Hunter J, Tsoli M, Ziegler DS, Dirksen U, Jager N, Balasubramanian GP, Kramm CM, Nathrath M, Bielack S, Baker SJ, Zhang J, McFarland JM, Getz G, Aguet F, Jabado N, Witt O, Pfister SM, Ligon KL, Hovestadt V, Kleinman CL, Long H, Jones DT, Bandopadhayay P, Phoenix TN. FOXR2 Is an Epigenetically Regulated Pan-Cancer Oncogene That Activates ETS Transcriptional Circuits. Cancer Res 2022; 82:2980-3001. [PMID: 35802025 PMCID: PMC9437574 DOI: 10.1158/0008-5472.can-22-0671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/11/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022]
Abstract
Forkhead box R2 (FOXR2) is a forkhead transcription factor located on the X chromosome whose expression is normally restricted to the testis. In this study, we performed a pan-cancer analysis of FOXR2 activation across more than 10,000 adult and pediatric cancer samples and found FOXR2 to be aberrantly upregulated in 70% of all cancer types and 8% of all individual tumors. The majority of tumors (78%) aberrantly expressed FOXR2 through a previously undescribed epigenetic mechanism that involves hypomethylation of a novel promoter, which was functionally validated as necessary for FOXR2 expression and proliferation in FOXR2-expressing cancer cells. FOXR2 promoted tumor growth across multiple cancer lineages and co-opted ETS family transcription circuits across cancers. Taken together, this study identifies FOXR2 as a potent and ubiquitous oncogene that is epigenetically activated across the majority of human cancers. The identification of hijacking of ETS transcription circuits by FOXR2 extends the mechanisms known to active ETS transcription factors and highlights how transcription factor families cooperate to enhance tumorigenesis. SIGNIFICANCE This work identifies a novel promoter that drives aberrant FOXR2 expression and delineates FOXR2 as a pan-cancer oncogene that specifically activates ETS transcriptional circuits across human cancers. See related commentary by Liu and Northcott, p. 2977.
Collapse
Affiliation(s)
- Jessica W. Tsai
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Paloma Cejas
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts, Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - Dayle K. Wang
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Smruti Patel
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David W. Wu
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Phonepasong Arounleut
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio
| | - Xin Wei
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio
| | - Ningxuan Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts, Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - Sudeepa Syamala
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts, Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - Frank P.B. Dubois
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alexander Crane
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kristine Pelton
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Jayne Vogelzang
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Cecilia Sousa
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Audrey Baguette
- Quantitative Life Sciences, McGill University, Montreal, Quebec H3A 2A7, Canada
- Lady Davis Research Institute, Jewish General Hospital, Montreal, Quebec H3T 1E2, Canada
| | - Xiaolong Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Alexandra L. Condurat
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Sarah E. Dixon-Clarke
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Biological Chemistry and Molecular Pharmacology, Boston, Massachusetts
| | - Kevin N. Zhou
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Sophie D. Lu
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Elizabeth M. Gonzalez
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Madison S. Chacon
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Jeromy J. Digiacomo
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Rushil Kumbhani
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Dana Novikov
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - J'Ya Hunter
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Maria Tsoli
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - David S. Ziegler
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Women's and Children's Health, University of New South Wales, Sydney, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Uta Dirksen
- West German Cancer Center, Pediatrics III, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Essen/Düsseldorf, Germany
| | - Natalie Jager
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg University Hospital and German Cancer Research Center (DKFZ) Heidelberg, Germany
- Division of Pediatric Neuro-Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Gnana Prakash Balasubramanian
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg University Hospital and German Cancer Research Center (DKFZ) Heidelberg, Germany
- Division of Pediatric Neuro-Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christof M. Kramm
- Division of Pediatric Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Michaela Nathrath
- Department of Pediatric Hematology and Oncology, Klinikum Kassel, Kassel, Germany
- Children's Cancer Research Centre and Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | | - Suzanne J. Baker
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
| | - François Aguet
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Nada Jabado
- Department of Human Genetics, McGill University, Montreal, H3A 0C7, Canada
- Department of Pediatrics, McGill University, and The Research Institute of the McGill University Health Centre, Montreal, H4A 3J1, Canada
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg University Hospital and German Cancer Research Center (DKFZ) Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, Immunology, and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Disease (NCT) Network, Germany
| | - Stefan M. Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg University Hospital and German Cancer Research Center (DKFZ) Heidelberg, Germany
- Division of Pediatric Neuro-Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, Immunology, and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Disease (NCT) Network, Germany
| | - Keith L. Ligon
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Volker Hovestadt
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Claudia L. Kleinman
- Lady Davis Research Institute, Jewish General Hospital, Montreal, Quebec H3T 1E2, Canada
- Department of Human Genetics, McGill University, Montreal, H3A 0C7, Canada
| | - Henry Long
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts, Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - David T.W. Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg University Hospital and German Cancer Research Center (DKFZ) Heidelberg, Germany
- Division of Pediatric Neuro-Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pratiti Bandopadhayay
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Timothy N. Phoenix
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
23
|
Almutairi MH, Alotaibi MM, Alonaizan R, Almutairi BO. Expression Profile of MAGE-B1 Gene and Its Hypomethylation Activation in Colon Cancer. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6066567. [PMID: 35937396 PMCID: PMC9348940 DOI: 10.1155/2022/6066567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 11/30/2022]
Abstract
Cancer-testis (CT) genes are typically expressed in the testes; however, they have been linked to aberrant expression in a variety of malignancies. MAGE-B family genes are an example of CT genes. Therefore, the overarching objective of this study was to examine the expressions of MAGE-B family genes in several patients with colon cancer (CC) to see if they might be employed as cancer biomarkers in the early phases of cancer detection and to improve treatment. In this investigation, RT-PCR was used to analyze MAGE-B family genes in neighboring normal colon (NC) tissue from 10 CC patients. In addition, the effect of DNA demethylation on the expression status of the MAGE-B1 gene was evaluated by RT-PCR in HCT116 and Caco-2 cells and by qRT-PCR for HCT116 only after treating both CC cell lines with varying concentrations of 5-aza-2'-deoxycytidine (1.0, 5.0, and 10.0 μM) for 48 or 72 hours. All MAGE-B family genes except for MAGE-B1 showed weak bands in several samples of NC tissues: MAGE-B2, MAGE-B3, MAGE-B4, MAGE-B5, and MAGE-B6 genes were observed in 40%, 50%, 40%, 30%, and 60% of the NC samples, respectively. Nonetheless, they had strong bands in multiple samples of CC tissues, with 70%, 90%, 60%, 50%, and 90% of the CC samples, respectively. Interestingly, MAGE-B1 was detected in 60% of CC tissues but not in NC tissues, suggesting that it is a potential biomarker for early CC detection. MAGE-B1 expression was not observed in either untreated or DMSO-treated HCT116 cells after 48 or 72 hours of treatment. However, according to the RT-PCR and qRT-PCR results, the MAGE-B1 gene was overexpressed in the HCT116 cells treated with three different concentrations of 5-aza-2'-deoxycytidine. This shows that demethylation plays a crucial role in MAGE-B1 expression activation.
Collapse
Affiliation(s)
- Mikhlid H. Almutairi
- Zoology Department, College of Science, King Saud University, P.O. Box: 2455, 11451 Riyadh, Saudi Arabia
| | - Mona M. Alotaibi
- Zoology Department, College of Science, King Saud University, P.O. Box: 2455, 11451 Riyadh, Saudi Arabia
| | - Rasha Alonaizan
- Zoology Department, College of Science, King Saud University, P.O. Box: 2455, 11451 Riyadh, Saudi Arabia
| | - Bader O. Almutairi
- Zoology Department, College of Science, King Saud University, P.O. Box: 2455, 11451 Riyadh, Saudi Arabia
| |
Collapse
|
24
|
Gupta R, Jit BP, Kumar S, Mittan S, Tanwer P, Ray MD, Mathur S, Perumal V, Kumar L, Rath GK, Sharma A. Leveraging epigenetics to enhance the efficacy of cancer-testis antigen: a potential candidate for immunotherapy. Epigenomics 2022; 14:865-886. [DOI: 10.2217/epi-2021-0479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological malignancy in women. The phenotype is characterized by delayed diagnosis, recurrence and drug resistance. Inherent immunogenicity potential, oncogenic function and expression of cancer-testis/germline antigen (CTA) in ovarian cancer render them a potential candidate for immunotherapy. Revolutionary clinical findings indicate that tumor antigen-mediated T-cell and dendritic cell-based immunotherapeutic approaches provide an excellent strategy for targeting tumors. Currently, dendritic cell vaccination for the treatment of B-cell lymphoma and CTA-based T-cell receptor transduced T-cell therapy involving MAGE-A4 and NY-ESO-1 are well documented and shown to be effective. This review highlighted the mechanical aspects of epigenetic drugs that can elicit a CTA-based humoral and cellular immune response and implicate T-cell and dendritic cell-based immunotherapeutic approaches.
Collapse
Affiliation(s)
- Rashmi Gupta
- Department of Biochemistry, National Cancer Institute – India, Jhajjar Campus, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Bimal Prasad Jit
- Department of Biochemistry, National Cancer Institute – India, Jhajjar Campus, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Santosh Kumar
- Department of Biochemistry, National Cancer Institute – India, Jhajjar Campus, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sandeep Mittan
- Montefiore Medical Center, Albert Einstein College of Medicine, NY 10467, USA
| | - Pranay Tanwer
- Laboratory Oncology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - M D Ray
- Department of Surgical Oncology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sandeep Mathur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Vanamail Perumal
- Department of Obstetrics & Gynecology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Lalit Kumar
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - G K Rath
- Department of Radiotherapy, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ashok Sharma
- Department of Biochemistry, National Cancer Institute – India, Jhajjar Campus, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| |
Collapse
|
25
|
Tuminello S, Zhang Y, Yang L, Durmus N, Snuderl M, Heguy A, Zeleniuch-Jacquotte A, Chen Y, Shao Y, Reibman J, Arslan AA. Global DNA Methylation Profiles in Peripheral Blood of WTC-Exposed Community Members with Breast Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19095104. [PMID: 35564499 PMCID: PMC9105091 DOI: 10.3390/ijerph19095104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 12/22/2022]
Abstract
Breast cancer represents the most common cancer diagnosis among World Trade Center (WTC)-exposed community members, residents, and cleanup workers enrolled in the WTC Environmental Health Center (WTC EHC). The primary aims of this study were (1) to compare blood DNA methylation profiles of WTC-exposed community members with breast cancer and WTC-unexposed pre-diagnostic breast cancer blood samples, and (2) to compare the DNA methylation differences among the WTC EHC breast cancer cases and WTC-exposed cancer-free controls. Gene pathway enrichment analyses were further conducted. There were significant differences in DNA methylation between WTC-exposed breast cancer cases and unexposed prediagnostic breast cancer cases. The top differentially methylated genes were Intraflagellar Transport 74 (IFT74), WD repeat-containing protein 90 (WDR90), and Oncomodulin (OCM), which are commonly upregulated in tumors. Probes associated with established tumor suppressor genes (ATM, BRCA1, PALB2, and TP53) were hypermethylated among WTC-exposed breast cancer cases compared to the unexposed group. When comparing WTC EHC breast cancer cases vs. cancer-free controls, there appeared to be global hypomethylation among WTC-exposed breast cancer cases compared to exposed controls. Functional pathway analysis revealed enrichment of several gene pathways in WTC-exposed breast cancer cases including endocytosis, proteoglycans in cancer, regulation of actin cytoskeleton, axon guidance, focal adhesion, calcium signaling, cGMP-PKG signaling, mTOR, Hippo, and oxytocin signaling. The results suggest potential epigenetic links between WTC exposure and breast cancer in local community members enrolled in the WTC EHC program.
Collapse
Affiliation(s)
- Stephanie Tuminello
- Department of Population Health, New York University Langone Health, New York, NY 10016, USA; (Y.Z.); (A.Z.-J.); (Y.C.); (Y.S.)
- Correspondence: (S.T.); (A.A.A.)
| | - Yian Zhang
- Department of Population Health, New York University Langone Health, New York, NY 10016, USA; (Y.Z.); (A.Z.-J.); (Y.C.); (Y.S.)
| | - Lei Yang
- Foundation Medicine, Cambridge, MA 02141, USA;
| | - Nedim Durmus
- Department of Medicine, New York University Langone Health, New York, NY 10016, USA; (N.D.); (J.R.)
| | - Matija Snuderl
- Department of Pathology, New York University Langone Health, New York, NY 10016, USA; (M.S.); (A.H.)
| | - Adriana Heguy
- Department of Pathology, New York University Langone Health, New York, NY 10016, USA; (M.S.); (A.H.)
- NYU Langone’s Genome Technology Center, New York, NY 10016, USA
| | - Anne Zeleniuch-Jacquotte
- Department of Population Health, New York University Langone Health, New York, NY 10016, USA; (Y.Z.); (A.Z.-J.); (Y.C.); (Y.S.)
- NYU Perlmutter Comprehensive Cancer Center, New York, NY 10016, USA
| | - Yu Chen
- Department of Population Health, New York University Langone Health, New York, NY 10016, USA; (Y.Z.); (A.Z.-J.); (Y.C.); (Y.S.)
- NYU Perlmutter Comprehensive Cancer Center, New York, NY 10016, USA
| | - Yongzhao Shao
- Department of Population Health, New York University Langone Health, New York, NY 10016, USA; (Y.Z.); (A.Z.-J.); (Y.C.); (Y.S.)
- NYU Perlmutter Comprehensive Cancer Center, New York, NY 10016, USA
| | - Joan Reibman
- Department of Medicine, New York University Langone Health, New York, NY 10016, USA; (N.D.); (J.R.)
| | - Alan A. Arslan
- Department of Population Health, New York University Langone Health, New York, NY 10016, USA; (Y.Z.); (A.Z.-J.); (Y.C.); (Y.S.)
- NYU Perlmutter Comprehensive Cancer Center, New York, NY 10016, USA
- Department of Obstetrics and Gynecology, New York University Langone Health, New York, NY 10016, USA
- Correspondence: (S.T.); (A.A.A.)
| |
Collapse
|
26
|
Liu B, Li G, Yang J, Li X, Wang H, Yang J, Wen H, He F. The mechanism of immune related signal pathway Egr2-FasL-Fas in transcription regulation and methylated modification of Paralichthys olivaceus under acute hypoxia stress. FISH & SHELLFISH IMMUNOLOGY 2022; 123:152-163. [PMID: 35219829 DOI: 10.1016/j.fsi.2022.02.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/04/2022] [Accepted: 02/18/2022] [Indexed: 06/14/2023]
Abstract
Apoptosis genes Egr2, Fas and FasL are related to immune responses. However, the mechanism of these genes inducing apoptosis in fish are still not very clear. An acute hypoxia treatment (1.73 ± 0.06 mg/L) for 24 h was carried out on Japanese flounder (Paralichthys olivaceus). The increasingly dense apoptotic signals at 3 h, 6 h, 12 h by TUNEL in skeletal muscle indicated that hypoxia could quickly affect muscle growth and development. Furthermore, we concluded that the Egr2-FasL-Fas signal pathway, which was located at the upstream of apoptotic executor protein caspases, was related to the apoptosis by quantitative real-time PCR, protein concentration detection in ELISA and double gene in situ hybridization methods. The mechanism of the pathway was researched in transcription regulation and epigenetic modification by dual-luciferase reporter assay and bisulfite modified method, respectively. Egr2, as a transcription factor, could up-regulate the expression of FasL gene. And its binding site was mainly between -479 to -1 of FasL gene promoter. The 5th CpG dinucleotides (-514) methylation levels in FasL gene were significantly affected by hypoxia, and they were negatively correlated with its expressions. These suggested that the -514 site may be a very important site to regulate the FasL gene expression. Above results, we concluded that hypoxia activated the immune related signal pathway Egr2-FasL-Fas to induced skeletal muscle apoptosis to affect growth and development of Japanese flounder. The study revealed the mechanism of hypoxia induced apoptosis, which could provide a reference for fish immunity and aquaculture management.
Collapse
Affiliation(s)
- Binghua Liu
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Guangling Li
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Jun Yang
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Xiaohui Li
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Hao Wang
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Jing Yang
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Haishen Wen
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China.
| | - Feng He
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China.
| |
Collapse
|
27
|
Lingg L, Rottenberg S, Francica P. Meiotic Genes and DNA Double Strand Break Repair in Cancer. Front Genet 2022; 13:831620. [PMID: 35251135 PMCID: PMC8895043 DOI: 10.3389/fgene.2022.831620] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/02/2022] [Indexed: 12/16/2022] Open
Abstract
Tumor cells show widespread genetic alterations that change the expression of genes driving tumor progression, including genes that maintain genomic integrity. In recent years, it has become clear that tumors frequently reactivate genes whose expression is typically restricted to germ cells. As germ cells have specialized pathways to facilitate the exchange of genetic information between homologous chromosomes, their aberrant regulation influences how cancer cells repair DNA double strand breaks (DSB). This drives genomic instability and affects the response of tumor cells to anticancer therapies. Since meiotic genes are usually transcriptionally repressed in somatic cells of healthy tissues, targeting aberrantly expressed meiotic genes may provide a unique opportunity to specifically kill cancer cells whilst sparing the non-transformed somatic cells. In this review, we highlight meiotic genes that have been reported to affect DSB repair in cancers derived from somatic cells. A better understanding of their mechanistic role in the context of homology-directed DNA repair in somatic cancers may provide useful insights to find novel vulnerabilities that can be targeted.
Collapse
Affiliation(s)
- Lea Lingg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Cancer Therapy Resistance Cluster, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Cancer Therapy Resistance Cluster, Department for BioMedical Research, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern, Bern, Switzerland
- *Correspondence: Sven Rottenberg, ; Paola Francica,
| | - Paola Francica
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Cancer Therapy Resistance Cluster, Department for BioMedical Research, University of Bern, Bern, Switzerland
- *Correspondence: Sven Rottenberg, ; Paola Francica,
| |
Collapse
|
28
|
Diacofotaki A, Loriot A, De Smet C. Identification of Tissue-Specific Gene Clusters Induced by DNA Demethylation in Lung Adenocarcinoma: More Than Germline Genes. Cancers (Basel) 2022; 14:cancers14041007. [PMID: 35205751 PMCID: PMC8870412 DOI: 10.3390/cancers14041007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/03/2022] [Accepted: 02/11/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Loss of DNA methylation is often observed in human tumors, but how this epigenetic alteration impacts the transcriptome of cancer cells remains largely undefined. So far, DNA hypomethylation in tumors has been associated with aberrant activation of a germline-specific gene expression program. Here, we exploited transcriptomic and methylomic datasets of lung adenocarcinoma to investigate the possibility that other gene expression programs also become ectopically activated in hypomethylated tumors. Remarkably, we found that DNA hypomethylation in lung adenocarcinoma is associated with ectopic activation of not only germline-specific genes, but also gene clusters displaying specific expression in the gastrointestinal tract, or in stratified epithelia. Interestingly, expression of genes in this latter group was of prognostic value. Together, our study brings novel insight into the transcriptomic changes associated with DNA hypomethylation in tumors, and is an incentive to explore the value of hypomethylated DNA sequences as cancer biomarkers. Abstract Genome-wide loss of DNA methylation is commonly observed in human cancers, but its impact on the tumor transcriptome remains ill-defined. Previous studies demonstrated that this epigenetic alteration causes aberrant activation of a germline-specific gene expression program. Here, we examined if DNA hypomethylation in tumors also leads to de-repression of gene clusters with other tissue specificities. To this end, we explored transcriptomic and methylomic datasets from human lung adenocarcinoma (LUAD) cell lines, normal lung, and lung alveolar type II cells, considered as the origin of LUAD. Interestingly, DNA demethylation in LUAD cell lines was associated with activation of not only germline-specific (CG) genes, but also gene clusters displaying specific expression in the gastrointestinal tract (GI), or in stratified epithelia (SE). Consistently, genes from all three clusters showed highly specific patterns of promoter methylation among normal tissues and cell types, and were generally sensitive to induction by a DNA demethylating agent. Analysis of TCGA datasets confirmed that demethylation and activation of CG, GI and SE genes also occurs in vivo in LUAD tumor tissues, in association with global genome hypomethylation. For genes of the GI cluster, we demonstrated that HNF4A is a necessary factor for transcriptional activation following promoter demethylation. Interestingly, expression of several SE genes, in particular FAM83A, correlated with both tumor grade and reduced patient survival. Together, our study uncovers novel cell-type specific gene clusters that become aberrantly activated in LUAD tumors in association with genome-wide hypomethylation.
Collapse
Affiliation(s)
- Anna Diacofotaki
- Group of Genetics and Epigenetics, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium; (A.D.); (A.L.)
| | - Axelle Loriot
- Group of Genetics and Epigenetics, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium; (A.D.); (A.L.)
- Group of Computational Biology and Bioinformatics, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Charles De Smet
- Group of Genetics and Epigenetics, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium; (A.D.); (A.L.)
- Correspondence:
| |
Collapse
|
29
|
Jin J, Tu J, Ren J, Cai Y, Chen W, Zhang L, Zhang Q, Zhu G. Comprehensive Analysis to Identify MAGEA3 Expression Correlated With Immune Infiltrates and Lymph Node Metastasis in Gastric Cancer. Front Oncol 2022; 11:784925. [PMID: 34970496 PMCID: PMC8712941 DOI: 10.3389/fonc.2021.784925] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022] Open
Abstract
Gastric cancer (GC) is an aggressive malignant tumor and causes a significant number of deaths every year. With the coming of the age of cancer immunotherapy, search for a new target in gastric cancer may benefit more advanced patients. Melanoma-associated antigen-A3 (MAGEA3), one of the members of the cancer-testis antigen (CTA) family, was considered an important part of cancer immunotherapy. We evaluate the potential role of MAGEA3 in GC through the TCGA database. The result revealed that MAGEA3 is upregulated in GC and linked to poor OS and lymph node metastasis. MAGEA3 was also correlated with immune checkpoints, TMB, and affected the tumor immune microenvironment and the prognosis of GC through CIBERSORT, TIMER, and Kaplan-Meier plotter database analysis. In addition, GSEA-identified MAGEA3 is involved in the immune regulation of GC. Moreover, the protein-protein interaction (PPI) networks of MAGEA3 were constructed through STRING database and MAGEA3-correlated miRNAs were screened based on the joint analysis of multiple databases. In terms of experimental verification, we constructed pET21a (+)/MAGEA3 restructuring plasmids and transformed to Escherichia coli Rosetta. MAGEA3 protein was used as an antigen after being expressed and purified and can effectively detect the specific IgG in 93 GC patients' serum specimens with 44.08% sensitivity and 92.54% specificity. Through further analysis, the positive rate of MAGEA3 was related to the stage and transfer number of lymph nodes. These results indicated that MAGEA3 is a novel biomarker and correlated with lymph node metastasis and immune infiltrates in GC, which could be a new target for immunotherapy.
Collapse
Affiliation(s)
- Jinji Jin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianxin Tu
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiahuan Ren
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiqi Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenjing Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lifang Zhang
- Department of Medical Microbiology and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Qiyu Zhang
- Department of Hepato-Bilio-Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guanbao Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
30
|
Sandhu S, Sou IF, Hunter JE, Salmon L, Wilson CL, Perkins ND, Hunter N, Davies OR, McClurg UL. Centrosome dysfunction associated with somatic expression of the synaptonemal complex protein TEX12. Commun Biol 2021; 4:1371. [PMID: 34880391 PMCID: PMC8654964 DOI: 10.1038/s42003-021-02887-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 11/12/2021] [Indexed: 12/22/2022] Open
Abstract
The synaptonemal complex (SC) is a supramolecular protein scaffold that mediates chromosome synapsis and facilitates crossing over during meiosis. In mammals, SC proteins are generally assumed to have no other function. Here, we show that SC protein TEX12 also localises to centrosomes during meiosis independently of chromosome synapsis. In somatic cells, ectopically expressed TEX12 similarly localises to centrosomes, where it is associated with centrosome amplification, a pathology correlated with cancer development. Indeed, TEX12 is identified as a cancer-testis antigen and proliferation of some cancer cells is TEX12-dependent. Moreover, somatic expression of TEX12 is aberrantly activated via retinoic acid signalling, which is commonly disregulated in cancer. Structure-function analysis reveals that phosphorylation of TEX12 on tyrosine 48 is important for centrosome amplification but not for recruitment of TEX12 to centrosomes. We conclude that TEX12 normally localises to meiotic centrosomes, but its misexpression in somatic cells can contribute to pathological amplification and dysfunction of centrosomes in cancers. Sandhu et al. report that the synaptonemal complex (SC) protein, TEX12, localises to centrosomes independently of the SC during meiosis. They also show that it provokes centrosome amplification in somatic cells, a pathology associated with cancer development.
Collapse
Affiliation(s)
- Sumit Sandhu
- Howard Hughes Medical Institute, Department of Microbiology and Molecular Genetics, University of California, Davis, CA, 95616, USA
| | - Ieng F Sou
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Jill E Hunter
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Lucy Salmon
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Caroline L Wilson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Neil D Perkins
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Neil Hunter
- Howard Hughes Medical Institute, Department of Microbiology and Molecular Genetics, University of California, Davis, CA, 95616, USA.
| | - Owen R Davies
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK. .,Wellcome Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh, EH9 3BF, UK.
| | - Urszula L McClurg
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK.
| |
Collapse
|
31
|
Wu SC, Münger K. Role and Clinical Utility of Cancer/Testis Antigens in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13225690. [PMID: 34830845 PMCID: PMC8616139 DOI: 10.3390/cancers13225690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/03/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022] Open
Abstract
Cancer/testis (CT) antigens exhibit selective expression predominantly in immunoprivileged tissues in non-pathological contexts but are aberrantly expressed in diverse cancers. Due to their expression pattern, they have historically been attractive targets for immunotherapies. A growing number of studies implicate CT antigens in almost all hallmarks of cancer, suggesting that they may act as cancer drivers. CT antigens are expressed in head and neck squamous cell carcinomas. However, their role in the pathogenesis of these cancers remains poorly studied. Given that CT antigens hold intriguing potential as therapeutic targets and as biomarkers for prognosis and that they can provide novel insights into oncogenic mechanisms, their further study in the context of head and squamous cell carcinoma is warranted.
Collapse
Affiliation(s)
- Sharon Changshan Wu
- Molecular Microbiology Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA;
| | - Karl Münger
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
- Correspondence:
| |
Collapse
|
32
|
Transcriptional overlap links DNA hypomethylation with DNA hypermethylation at adjacent promoters in cancer. Sci Rep 2021; 11:17346. [PMID: 34462486 PMCID: PMC8405634 DOI: 10.1038/s41598-021-96844-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/17/2021] [Indexed: 12/15/2022] Open
Abstract
Tumor development involves alterations in DNA methylation patterns, which include both gains (hypermethylation) and losses (hypomethylation) in different genomic regions. The mechanisms underlying these two opposite, yet co-existing, alterations in tumors remain unclear. While studying the human MAGEA6/GABRA3 gene locus, we observed that DNA hypomethylation in tumor cells can lead to the activation of a long transcript (CT-GABRA3) that overlaps downstream promoters (GABRQ and GABRA3) and triggers their hypermethylation. Overlapped promoters displayed increases in H3K36me3, a histone mark deposited during transcriptional elongation and known to stimulate de novo DNA methylation. Consistent with such a processive mechanism, increases in H3K36me3 and DNA methylation were observed over the entire region covered by the CT-GABRA3 overlapping transcript. Importantly, experimental induction of CT-GABRA3 by depletion of DNMT1 DNA methyltransferase, resulted in a similar pattern of regional DNA hypermethylation. Bioinformatics analyses in lung cancer datasets identified other genomic loci displaying this process of coupled DNA hypo/hypermethylation, and some of these included tumor suppressor genes, e.g. RERG and PTPRO. Together, our work reveals that focal DNA hypomethylation in tumors can indirectly contribute to hypermethylation of nearby promoters through activation of overlapping transcription, and establishes therefore an unsuspected connection between these two opposite epigenetic alterations.
Collapse
|
33
|
Achinko DA, Dormer A, Narayanan M, Norman EF. Targeted immune epitope prediction to HHLA2 and MAGEB5 protein variants as therapeutic approach to related viral diseases. BMC Immunol 2021; 22:49. [PMID: 34320928 PMCID: PMC8316541 DOI: 10.1186/s12865-021-00440-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 07/20/2021] [Indexed: 12/28/2022] Open
Abstract
Background Targeted immunotherapy is mostly associated with cancer treatment wherein designed molecules engage signaling pathways and mutant proteins critical to the survival of the cell. One of several genetic approaches is the use of in silico methods to develop immune epitopes targeting specific antigenic regions on related mutant proteins. In a recent study we showed a functional association between the gamma retrovirus HERV-H Long Terminal Associating (HHLA1, HHLA2 and HHLA3) proteins and melanoma associated antigen of the B class proteins (MAGEB5), with a resultant decrease in expression of HLA class I and II immune variants. HLA-C and HLA-DRB5 were the main HLA class I and II Immune variants, respectively, that showed expression changes across viral samples of interest. Specific immune variants for HLA-C and HLA-DRB5 were filtered for the top ten based on their relative frequency of counts across the samples. Results Protein variants for HHLA1, HHLA2, HHLA3 and MAGEB5 were used to predict antigenic epitope peptides to immune peptide-MHC class I and II binding using artificial neural networks. For IC50 peptide scores (PS) ≥ 0.5 with a transformed binding ability between 0 and 1, the top 5 epitopes identified for all targeted genes HHLA1,2 & 3 and MAGEB5 were qualified as strong or weak binders according to the threshold. Domain analysis using NCBI Conserved Domain Database (CDD) identified HHLA2 with immunoglobulin-like domains (Ig_C1-set) and MAGEB5 with the MAGE Homology Domain (MHD). Linear regression showed a statistical correlation (P < 0.001) for HHLA2 and MAGEB5 predicted epitope peptides to HLA-C but not HLA-DRB5. The prediction model identified HLA-C variant 9 (HLA-C9, BAA08825.1 HLA-B*1511) at 1.1% as the most valuable immune target for clinical considerations. Identification of the 9-mer epitope peptide within the domain showed for HHLA2: YANRTSLFY (PS = 0.5837) and VLAYYLSSSQNTIIN (PS = 0.77) for HLA-C and HLA-DRB5, respectively and for MAGEB5, peptides: FVRLTYLEY (PS = 0.5293) and YPAHYQFLWGPRAYT (PS = 0.62) for HLA-C and HLA-DRB5, respectively. Conclusion Specific immune responses to targeted epitope peptides and their prediction models, suggested co-expression and co-evolution for HHLA2 and MAGEB5 in viral related diseases. HHLA2 and MAGEB5 could be considered markers for virus related tumors and targeted therapy for oncogenic diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-021-00440-w.
Collapse
Affiliation(s)
- Daniel A Achinko
- PepVax, Inc., 0411 Motor City Drive, Suite #750, Bethesda, MD, 20817, USA.
| | - Anton Dormer
- PepVax, Inc., 0411 Motor City Drive, Suite #750, Bethesda, MD, 20817, USA
| | - Mahesh Narayanan
- PepVax, Inc., 0411 Motor City Drive, Suite #750, Bethesda, MD, 20817, USA
| | - Elton F Norman
- PepVax, Inc., 0411 Motor City Drive, Suite #750, Bethesda, MD, 20817, USA
| |
Collapse
|
34
|
Oncogenic activity and cellular functionality of melanoma associated antigen A3. Biochem Pharmacol 2021; 192:114700. [PMID: 34303709 DOI: 10.1016/j.bcp.2021.114700] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022]
Abstract
Cancer testis antigen Melanoma associated antigen A3 (MAGE-A3) has been subject of research for many years. Being expressed in various tumor types and influencing proliferation, metastasis, and tumor pathogenicity, MAGE-A3 is an attractive target for cancer therapy, particularly because in healthy tissues, MAGE-A3 is only expressed in testes and placenta. MAGE-A3 acts as a cellular master regulator by stimulating E3 ubiquitin ligase tripartite motif-containing protein 28 (TRIM28), resulting in regulation of various cellular targets. These include tumor suppressor protein p53 and cellular energy sensor AMP-activated protein kinase (AMPK). The restricted expression of MAGE-A3 in tumor cells makes MAGE-A3 an attractive target for vaccine-based immune therapy. However, although phase I and phase II clinical trials involving MAGE-A3-specific immunotherapeutic interventions were promising, large phase III studies failed. This article gives an overview about the role of MAGE-A3 as a cellular master switch and discusses approaches to improve MAGE-A3-based immunotherapies.
Collapse
|
35
|
Wang X, Zhou S, Chu C, Yang M, Huo D, Hou C. Dual Methylation-Sensitive Restriction Endonucleases Coupling with an RPA-Assisted CRISPR/Cas13a System (DESCS) for Highly Sensitive Analysis of DNA Methylation and Its Application for Point-of-Care Detection. ACS Sens 2021; 6:2419-2428. [PMID: 34019391 DOI: 10.1021/acssensors.1c00674] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
High-performance detection of DNA methylation possesses great significance for the diagnosis and therapy of cancer. Herein, for the first time, we present a digestion strategy based on dual methylation-sensitive restriction endonucleases coupling with a recombinase polymerase amplification (RPA)-assisted CRISPR/Cas13a system (DESCS) for accurate and sensitive determination of site-specific DNA methylation. This dual methylation-sensitive restriction endonuclease system selectively digests the unmethylated target but exhibits no response to methylated DNA. Therefore, the intact methylated DNA target triggers the RPA reaction for rapid signal amplification. In contrast, the digested unmethylated target initiates no RPA reaction. RPA products with a T7 promoter can execute the T7 transcription in the presence of T7 RNA polymerase to generate a large number of single-stranded RNA (ssRNA). This ssRNA can be recognized by CRISPR/Cas13a to induce the ssRNase activity of Cas13a, showing the indiscriminate cleavage of the collateral FQ reporter to release the fluorescence signal. With such a design, by combining the unique features of dual methylation-sensitive restriction endonucleases with RPA-assisted CRISPR/Cas13a, the DESCS system not only presents the rapid and powerful signal amplification for the determination of methylated DNA with ultrahigh sensitivity but also effectively eliminates the false positive influences from incomplete digestion of the unmethylated target. More importantly, 0.01% methylation level can be effectively distinguished with the existence of excess unmethylated DNA. In addition, the DESCS assay is integrated into the lateral flow biosensor (LFB) for the point-of-care determination of DNA methylation. In view of the superiorities in high sensitivity, outstanding selectivity, and ease of operation, the DESCS system will provide a reliable assay for site-specific analysis of methylation.
Collapse
Affiliation(s)
- Xianfeng Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - Shiying Zhou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - Chengxiang Chu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - Mei Yang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - Danqun Huo
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
- Chongqing Key Laboratory of Bio-perception & Intelligent Information Processing, School of Microelectronics and Communication Engineering, Chongqing University, Chongqing 400044, P. R. China
| | - Changjun Hou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
- Chongqing Key Laboratory of Bio-perception & Intelligent Information Processing, School of Microelectronics and Communication Engineering, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
36
|
Meng X, Sun X, Liu Z, He Y. A novel era of cancer/testis antigen in cancer immunotherapy. Int Immunopharmacol 2021; 98:107889. [PMID: 34174699 DOI: 10.1016/j.intimp.2021.107889] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/24/2022]
Abstract
Immunotherapy is a regimen that is especially utilized in many advanced cancers. Tumor antigens include tumor-specific antigens and tumor-associated antigens, and they function as targets for immunotherapy, such as cancer vaccines and autologous T cells. Cancer/testis antigens (CTAs), which is a group of genes that are restrictedly expressed in malignant cells as well as some germline cells, are tumor-associated antigens. These expression characteristics make CTAs promising candidates for vaccine or T cell therapy targets. Cancer vaccines utilize cancer antigens to induce specific cellular and humoral immune responses to strengthen the body's immune system. T cell transfer therapy refers to genetically modifying T cells to express antigen-specific T cell receptors or chimeric antigen receptors, both of which can be directly activated by tumor antigens. Moreover, combined therapies are being investigated based on CTAs. Current studies have mainly focused on MAGE-A, NY-ESO-1, and IL-13Rα. And we will review clinical trials of CTA-based immunotherapies related to these three antigens. We will summarize completed trials and results and examine the future trends in immunotherapy.
Collapse
Affiliation(s)
- Xiaoyan Meng
- Department of Oral Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai 200011, China
| | - Xueqing Sun
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhonglong Liu
- Department of Oral Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai 200011, China
| | - Yue He
- Department of Oral Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai 200011, China.
| |
Collapse
|
37
|
Chen C, Gao D, Huo J, Qu R, Guo Y, Hu X, Luo L. Multiomics analysis reveals CT83 is the most specific gene for triple negative breast cancer and its hypomethylation is oncogenic in breast cancer. Sci Rep 2021; 11:12172. [PMID: 34108519 PMCID: PMC8190062 DOI: 10.1038/s41598-021-91290-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 05/25/2021] [Indexed: 02/05/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer (BrC) subtype lacking effective therapeutic targets currently. The development of multi-omics databases facilities the identification of core genes for TNBC. Using TCGA-BRCA and METABRIC datasets, we identified CT83 as the most TNBC-specific gene. By further integrating FUSCC-TNBC, CCLE, TCGA pan-cancer, Expression Atlas, and Human Protein Atlas datasets, we found CT83 is frequently activated in TNBC and many other cancers, while it is always silenced in non-TNBC, 120 types of normal non-testis tissues, and 18 types of blood cells. Notably, according to the TCGA-BRCA methylation data, hypomethylation on chromosome X 116,463,019 to 116,463,039 is significantly correlated with the abnormal activation of CT83 in BrC. Using Kaplan-Meier Plotter, we demonstrated that activated CT83 is significantly associated with unfavorably overall survival in BrC and worse outcomes in some other cancers. Furthermore, GSEA suggested that the abnormal activation of CT83 in BrC is probably oncogenic by triggering the activation of cell cycle signaling. Meanwhile, we also noticed copy number variations and mutations of CT83 are quite rare in any cancer type, and its role in immune infiltration is not significant. In summary, we highlighted the significance of CT83 for TNBC and presented a comprehensive bioinformatics strategy for single-gene analysis in cancer.
Collapse
Affiliation(s)
- Chen Chen
- grid.452884.7Breast and Thyroid Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, Zunyi, 563000 Guizhou China
| | - Dan Gao
- grid.452884.7Breast and Thyroid Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, Zunyi, 563000 Guizhou China
| | - Jinlong Huo
- grid.452884.7Breast and Thyroid Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, Zunyi, 563000 Guizhou China
| | - Rui Qu
- grid.452884.7Breast and Thyroid Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, Zunyi, 563000 Guizhou China
| | - Youming Guo
- grid.452884.7Breast and Thyroid Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, Zunyi, 563000 Guizhou China
| | - Xiaochi Hu
- grid.452884.7Breast and Thyroid Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, Zunyi, 563000 Guizhou China
| | - Libo Luo
- grid.452884.7Breast and Thyroid Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Fenghuang N Rd, Zunyi, 563000 Guizhou China
| |
Collapse
|
38
|
Shang Y, Jiang YL, Ye LJ, Chen LN, Ke Y. Resveratrol acts via melanoma-associated antigen A12 (MAGEA12)/protein kinase B (Akt) signaling to inhibit the proliferation of oral squamous cell carcinoma cells. Bioengineered 2021; 12:2253-2262. [PMID: 34085601 PMCID: PMC8806796 DOI: 10.1080/21655979.2021.1934242] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The present study examined how resveratrol affects cell growth and MAGEA12/Akt signaling pathway in OSCC cells. Cal-27 cells were transiently transfected with a plasmid encoding MAGEA12, and the effects of overexpression were assessed in terms of cell viability, colony formation and the epithelial–mesenchymal transition. Cal-27 cells and MAGEA12-overexpressing cells were treated with resveratrol, then the cell viability and colony formation were also assessed by CCK8 assay and microscope, respectively. Levels of MAGEA12, p-Akt, Akt, Cyclin D1, and CDK14 genes and these proteins were analyzed using quantitative reverse-transcription polymerase-chain reaction and western blot. In the present research, we first generated and transiently transfected MAGEA12 plasmid into Cal-27 cells. Our results suggested that overexpressing MAGEA12 led to an increase in levels of phospho-Akt, which was associated with increased cell viability, colony formation. Moreover, overexpressing MAGEA12 also resulted in the up-regulation of Cyclin D1 and CDK14, indicating MAGEA12 induces the cell proliferation of Cal-27 cells. In addition, these effects were partially reversed by inhibiting Akt. Furthermore, resveratrol could inhibit the proliferation and colony in Cal-27 cells and decrease the expressions of MAGEA12 and p-Akt depending on the time and concentration. These effects were also partially reversed by MAGEA12 overexpression and Akt activation. In summary, resveratrol may suppress the growth of OSCC cells by inactivating MAGEA12/Akt signaling. These findings suggest that resveratrol may be a therapeutic drug for OSCC in clinical.
Collapse
Affiliation(s)
- Yu Shang
- Department of Stomatology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, P.R.C
| | - Yu-Ling Jiang
- Department of Stomatology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, P.R.C
| | - Li-Jun Ye
- Department of Stomatology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, P.R.C
| | - Li-Na Chen
- Department of Stomatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, P.R.C
| | - Yue Ke
- Department of Stomatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, P.R.C
| |
Collapse
|
39
|
Wu M, Wang S, Chen JY, Zhou LJ, Guo ZW, Li YH. Therapeutic cancer vaccine therapy for acute myeloid leukemia. Immunotherapy 2021; 13:863-877. [PMID: 33955237 DOI: 10.2217/imt-2020-0277] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Antitumor function of the immune system has been harnessed to eradicate tumor cells as cancer therapy. Therapeutic cancer vaccines aim to help immune cells recognize tumor cells, which are difficult to target owing to immune escape. Many attempts at vaccine designs have been conducted throughout the last decades. In addition, as the advanced understanding of immunosuppressive mechanisms mediated by tumor cells, combining cancer vaccines with other immune therapies seems to be more efficient for cancer treatment. Acute myeloid leukemia (AML) is the most common acute leukemia in adults with poor prognosis. Evidence has shown T-cell-mediated immune responses in AML, which encourages the utility of immune therapies in AML. This review discusses cancer vaccines in AML from vaccine design as well as recent progress in vaccination combination with other immune therapies.
Collapse
Affiliation(s)
- Ming Wu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.,Department of Hematology, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Sheng Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jian-Yu Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Li-Juan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Zi-Wen Guo
- Department of Hematology, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Yu-Hua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
40
|
Fan C, Qu H, Wang X, Sobhani N, Wang L, Liu S, Xiong W, Zeng Z, Li Y. Cancer/testis antigens: from serology to mRNA cancer vaccine. Semin Cancer Biol 2021; 76:218-231. [PMID: 33910064 DOI: 10.1016/j.semcancer.2021.04.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/01/2023]
Abstract
Cancer/testis antigens (CTAs) are a group of tumor antigens expressed in numerous cancer tissues, as well as in the testis and placental tissues. There are over 200 CTAs supported by serology and expression data. The expression patterns of CTAs reflect the similarities between the processes of gametogenesis and tumorigenesis. It is notable that CTAs are highly expressed in three types of cancers (lung cancer, bladder cancer, and skin cancer), all of which have a metal etiology. Here, we review the expression, regulation, and function of CTAs and their translational prospects as cancer biomarkers and treatment targets. Many CTAs are highly immunogenic, tissue-specific, and frequently expressed in cancer tissues but not under physiological conditions, rendering them promising candidates for cancer detection. Some CTAs are associated with clinical outcomes, so they may serve as prognostic biomarkers. A small number of CTAs are membrane-bound, making them ideal targets for chimeric antigen receptor (CAR) T cells. Mounting evidence suggests that CTAs induce humoral or cellular immune responses, providing cancer immunotherapeutic opportunities for T-cell receptors (TCRs), CAR T cell, antibody-based therapy and peptide- or mRNA-based vaccines. Indeed, CTAs are the dominating non-mutated targets in mRNA cancer vaccine development. Clinical trials on CTA TCR and vaccines have shown effectiveness, safety, and tolerance, but these successes are limited to a small number of patients. In-depth studies on CTA expression and function are needed to improve CTA-based immunotherapy.
Collapse
Affiliation(s)
- Chunmei Fan
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China; Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Hongke Qu
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Xu Wang
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Navid Sobhani
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Leiming Wang
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Shuanglin Liu
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China.
| | - Yong Li
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States.
| |
Collapse
|
41
|
Jakobsen MK, Traynor S, Stæhr M, Duijf PG, Nielsen AY, Terp MG, Pedersen CB, Guldberg P, Ditzel HJ, Gjerstorff MF. The Cancer/Testis Antigen Gene VCX2 Is Rarely Expressed in Malignancies but Can Be Epigenetically Activated Using DNA Methyltransferase and Histone Deacetylase Inhibitors. Front Oncol 2021; 10:584024. [PMID: 33634013 PMCID: PMC7900521 DOI: 10.3389/fonc.2020.584024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/14/2020] [Indexed: 01/25/2023] Open
Abstract
Identification of novel tumor-specific targets is important for the future development of immunotherapeutic strategies using genetically engineered T cells or vaccines. In this study, we characterized the expression of VCX2, a member of the VCX/Y cancer/testis antigen family, in a large panel of normal tissues and tumors from multiple cancer types using immunohistochemical staining and RNA expression data. In normal tissues, VCX2 was detected in the germ cells of the testis at all stages of maturation but not in any somatic tissues. Among malignancies, VCX2 was only found in tumors of a small subset of melanoma patients and thus rarely expressed compared to other cancer/testis antigens such as GAGE and MAGE-A. The expression of VCX2 correlated with that of other VCX/Y genes. Importantly, we found that expression of VCX2 was inversely correlated with promoter methylation and could be activated by treatment with a DNA methyltransferase inhibitor in multiple breast cancer and melanoma cell lines and a breast cancer patient-derived xenograft. The effect could be further potentiated by combining the DNA methyltransferase inhibitor with a histone deacetylase inhibitor. Our results show that the expression of VCX2 can be epigenetically induced in cancer cells and therefore could be an attractive target for immunotherapy of cancer.
Collapse
Affiliation(s)
- Mie K Jakobsen
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sofie Traynor
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Mette Stæhr
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Pascal G Duijf
- Institute of Health and Biomedical Innovation, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Aaraby Y Nielsen
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Mikkel G Terp
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Christina B Pedersen
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Per Guldberg
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Molecular Diagnostics Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Henrik J Ditzel
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark.,Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark.,Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| |
Collapse
|
42
|
Liu C, Huang R, Wang L, Liang G. Functional Identification of EjGIF1 in Arabidopsis and Preliminary Analysis of Its Regulatory Mechanisms in the Formation of Triploid Loquat Leaf Heterosis. FRONTIERS IN PLANT SCIENCE 2021; 11:612055. [PMID: 33510754 PMCID: PMC7835675 DOI: 10.3389/fpls.2020.612055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/11/2020] [Indexed: 06/12/2023]
Abstract
Although several results have been obtained in triploid loquat heterosis (i.e., leaf size of triploid loquat) studies in the past years, the underlying mechanisms of the heterosis are still largely unknown, especially the regulation effects of one specific gene on the corresponding morphology heterosis. In this study, we sought to further illustrate the regulatory mechanisms of one specific gene on the leaf size heterosis of triploid loquats. A leaf size development-related gene (EjGIF1) and its promoter were successfully cloned. Ectopic expression of EjGIF1 in Arabidopsis showed that the leaf size of transgenic plantlets was larger than that of WTs, and the transgenic plantlets had more leaves than WTs. Quantitative Reverse Transcription PCR (qRT-PCR) showed that the expression level of EjGIF1 showed an AHP expression pattern in most of the hybrids, and this was consistent with our previous phenotype observations. Structure analysis of EjGIF1 promoter showed that there were significantly more light-responsive elements than other elements. To further ascertain the regulatory mechanisms of EjGIF1 on triploid loquat heterosis, the methylation levels of EjGIF1 promoter in different ploidy loquats were analyzed by using bisulfite sequencing. Surprisingly, the total methylation levels of EjGIF1 promoter in triploid showed a decreasing trend compared with the mid-parent value (MPV), and this was also consistent with the qRT-PCR results of EjGIF1. Taken together, our results suggested that EjGIF1 played an important role in promoting leaf size development of loquat, and demethylation of EjGIF1 promoter in triploid loquats caused EjGIF1 to exhibit over-dominance expression pattern and then further to promote leaf heterosis formation. In conclusion, EjGIF1 played an important role in the formation of triploid loquat leaf size heterosis.
Collapse
Affiliation(s)
- Chao Liu
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Renwei Huang
- Sichuan Provincial Key Laboratory for Development and Utilization of Characteristic Horticultural Biological Resources, College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Lingli Wang
- Technical Advice Station of Economic Crop, Chongqing, China
| | - Guolu Liang
- College of Horticulture and Landscape Architecture, Southwest University, Chongqing, China
| |
Collapse
|
43
|
Florke Gee RR, Chen H, Lee AK, Daly CA, Wilander BA, Fon Tacer K, Potts PR. Emerging roles of the MAGE protein family in stress response pathways. J Biol Chem 2020; 295:16121-16155. [PMID: 32921631 PMCID: PMC7681028 DOI: 10.1074/jbc.rev120.008029] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/08/2020] [Indexed: 12/21/2022] Open
Abstract
The melanoma antigen (MAGE) proteins all contain a MAGE homology domain. MAGE genes are conserved in all eukaryotes and have expanded from a single gene in lower eukaryotes to ∼40 genes in humans and mice. Whereas some MAGEs are ubiquitously expressed in tissues, others are expressed in only germ cells with aberrant reactivation in multiple cancers. Much of the initial research on MAGEs focused on exploiting their antigenicity and restricted expression pattern to target them with cancer immunotherapy. Beyond their potential clinical application and role in tumorigenesis, recent studies have shown that MAGE proteins regulate diverse cellular and developmental pathways, implicating them in many diseases besides cancer, including lung, renal, and neurodevelopmental disorders. At the molecular level, many MAGEs bind to E3 RING ubiquitin ligases and, thus, regulate their substrate specificity, ligase activity, and subcellular localization. On a broader scale, the MAGE genes likely expanded in eutherian mammals to protect the germline from environmental stress and aid in stress adaptation, and this stress tolerance may explain why many cancers aberrantly express MAGEs Here, we present an updated, comprehensive review on the MAGE family that highlights general characteristics, emphasizes recent comparative studies in mice, and describes the diverse functions exerted by individual MAGEs.
Collapse
Affiliation(s)
- Rebecca R Florke Gee
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Helen Chen
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Anna K Lee
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Christina A Daly
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Benjamin A Wilander
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Klementina Fon Tacer
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; School of Veterinary Medicine, Texas Tech University, Amarillo, Texas, USA.
| | - Patrick Ryan Potts
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
| |
Collapse
|
44
|
Leko V, Rosenberg SA. Identifying and Targeting Human Tumor Antigens for T Cell-Based Immunotherapy of Solid Tumors. Cancer Cell 2020; 38:454-472. [PMID: 32822573 PMCID: PMC7737225 DOI: 10.1016/j.ccell.2020.07.013] [Citation(s) in RCA: 268] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 12/20/2022]
Abstract
Cancer elimination in humans can be achieved with immunotherapy that relies on T lymphocyte-mediated recognition of tumor antigens. Several types of these antigens have been recognized based on their cellular origins and expression patterns, while their detection has been greatly facilitated by recent achievements in next-generation sequencing and immunopeptidomics. Some of them have been targeted in clinical trials with various immunotherapy approaches, while many others remain untested. Here, we discuss molecular identification of different tumor antigen types, and the clinical safety and efficacy of targeting them with immunotherapy. Additionally, we suggest strategies to increase the efficacy and availability of antigen-directed immunotherapies for treatment of patients with metastatic cancer.
Collapse
Affiliation(s)
- Vid Leko
- Surgery Branch, National Cancer Institute, National Institutes of Health, Building 10-CRC, Room 3-3942, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Steven A Rosenberg
- Surgery Branch, National Cancer Institute, National Institutes of Health, Building 10-CRC, Room 3-3942, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
45
|
Poojary M, Jishnu PV, Kabekkodu SP. Prognostic Value of Melanoma-Associated Antigen-A (MAGE-A) Gene Expression in Various Human Cancers: A Systematic Review and Meta-analysis of 7428 Patients and 44 Studies. Mol Diagn Ther 2020; 24:537-555. [PMID: 32548799 PMCID: PMC7497308 DOI: 10.1007/s40291-020-00476-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Members of the melanoma-associated antigen-A (MAGE-A) subfamily are overexpressed in many cancers and can drive cancer progression, metastasis, and therapeutic recurrence. OBJECTIVE This study is the first comprehensive meta-analysis evaluating the prognostic utility of MAGE-A members in different cancers. METHODS A systematic literature search was conducted in PubMed, Google Scholar, Science Direct, and Web of Science. The pooled hazard ratios with 95% confidence intervals were estimated to evaluate the prognostic significance of MAGE-A expression in various cancers. RESULTS In total, 44 eligible studies consisting of 7428 patients from 11 countries were analysed. Univariate and multivariate analysis for overall survival, progression-free survival, and disease-free survival showed a significant association between high MAGE-A expression and various cancers (P < 0.00001). Additionally, subgroup analysis demonstrated that high MAGE-A expression was significantly associated with poor prognosis for lung, gastrointestinal, breast, and ovarian cancer in both univariate and multivariate analysis for overall survival. CONCLUSION Overexpression of MAGE-A subfamily members is linked to poor prognosis in multiple cancers. Therefore, it could serve as a potential prognostic marker of poor prognosis in cancers.
Collapse
Affiliation(s)
- Manish Poojary
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Padacherri Vethil Jishnu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India.
| |
Collapse
|
46
|
Cusack M, King HW, Spingardi P, Kessler BM, Klose RJ, Kriaucionis S. Distinct contributions of DNA methylation and histone acetylation to the genomic occupancy of transcription factors. Genome Res 2020; 30:1393-1406. [PMID: 32963030 PMCID: PMC7605266 DOI: 10.1101/gr.257576.119] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 08/21/2020] [Indexed: 12/12/2022]
Abstract
Epigenetic modifications on chromatin play important roles in regulating gene expression. Although chromatin states are often governed by multilayered structure, how individual pathways contribute to gene expression remains poorly understood. For example, DNA methylation is known to regulate transcription factor binding but also to recruit methyl-CpG binding proteins that affect chromatin structure through the activity of histone deacetylase complexes (HDACs). Both of these mechanisms can potentially affect gene expression, but the importance of each, and whether these activities are integrated to achieve appropriate gene regulation, remains largely unknown. To address this important question, we measured gene expression, chromatin accessibility, and transcription factor occupancy in wild-type or DNA methylation-deficient mouse embryonic stem cells following HDAC inhibition. We observe widespread increases in chromatin accessibility at retrotransposons when HDACs are inhibited, and this is magnified when cells also lack DNA methylation. A subset of these elements has elevated binding of the YY1 and GABPA transcription factors and increased expression. The pronounced additive effect of HDAC inhibition in DNA methylation-deficient cells demonstrates that DNA methylation and histone deacetylation act largely independently to suppress transcription factor binding and gene expression.
Collapse
Affiliation(s)
- Martin Cusack
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Hamish W King
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, United Kingdom
| | - Paolo Spingardi
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Benedikt M Kessler
- Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ, United Kingdom
| | - Robert J Klose
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, United Kingdom
| | - Skirmantas Kriaucionis
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, OX3 7DQ, United Kingdom;
| |
Collapse
|
47
|
Jakobsen MK, Gjerstorff MF. CAR T-Cell Cancer Therapy Targeting Surface Cancer/Testis Antigens. Front Immunol 2020; 11:1568. [PMID: 32983080 PMCID: PMC7492268 DOI: 10.3389/fimmu.2020.01568] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Affiliation(s)
- Mie K Jakobsen
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark.,Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| |
Collapse
|
48
|
Zhong G, Jin G, Zeng W, Yu C, Li Y, Zhou J, Zhang L, Yu L. Conjugation of TLR7 Agonist Combined with Demethylation Treatment Improves Whole-Cell Tumor Vaccine Potency in Acute Myeloid Leukemia. Int J Med Sci 2020; 17:2346-2356. [PMID: 32922200 PMCID: PMC7484644 DOI: 10.7150/ijms.49983] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/09/2020] [Indexed: 11/06/2022] Open
Abstract
Background: Acute myeloid leukemia (AML) is a malignant hematological disease with high refractory rate. Immune escape of AML cells is one of the underlying mechanisms mediating the relapse of the cancers. Various immunotherapies based on the 'patients' immune response to tumor cells have been developed to targeting the immune escape of AML cells, which lead to the minimal residual disease (MRD) after treatment. But the efficacy of those treatments or the combination of treatments remains unsatisfactory. Methods: A Toll-like receptor (TLR)-7 agonist SZU-106 was chemically synthesized. SZU-106 was conjugated to Decitabine (DAC), a demethylation agent, treated AML cells to construct tumor vaccine. The potency of the newly constructed AML cell vaccine, SZU-106-DAC-AML was tested in vitro and in vivo for the expression of tumor antigens and the activation level of immune responses. Results: Compared to the well characterized TLR7 agonist R848, SZU-106 has a comparable potency to activate TLR7 signaling pathway. SZU-106-DAC-AML, constructed by conjugating SZU-106 to DAC treated tumor cells, exhibited increased expression of tumor antigens, and enhanced the activation of DC cells and T cells in vitro and in vivo. The result of xenograft tumor model showed that SZU-106-DAC-AML tumor vaccine greatly inhibited tumor growth and prolonged animal survival. Conclusions: Conjugation of TLR7 agonist combined with up-regulation of tumor antigen expression improved the effectiveness of whole-cell tumor vaccine in AML.
Collapse
Affiliation(s)
- Guocheng Zhong
- Department of Hematology and BMT center, the first Medical Center, Chinese PLA General Hospital. Beijing 100853, China
- Department of Hematology and Oncology, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Guangyi Jin
- Carson International Cancer Center, Department of Medicine, Shenzhen University, Shenzhen 518037, China
| | - Wei Zeng
- Department of Hematology and Oncology, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Changhua Yu
- Department of Hematology and Oncology, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Yan Li
- Department of Hematology and BMT center, the first Medical Center, Chinese PLA General Hospital. Beijing 100853, China
| | - Ji Zhou
- Carson International Cancer Center, Department of Medicine, Shenzhen University, Shenzhen 518037, China
| | - Li Zhang
- Carson International Cancer Center, Department of Medicine, Shenzhen University, Shenzhen 518037, China
| | - Li Yu
- Department of Hematology and BMT center, the first Medical Center, Chinese PLA General Hospital. Beijing 100853, China
- Department of Hematology and Oncology, Shenzhen University General Hospital, Shenzhen 518055, China
| |
Collapse
|
49
|
Kutilin DS. Regulation of Gene Expression of Cancer/Testis Antigens in Colorectal Cancer Patients. Mol Biol 2020. [DOI: 10.1134/s0026893320040093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
50
|
Arslan AA, Tuminello S, Yang L, Zhang Y, Durmus N, Snuderl M, Heguy A, Zeleniuch-Jacquotte A, Shao Y, Reibman J. Genome-Wide DNA Methylation Profiles in Community Members Exposed to the World Trade Center Disaster. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17155493. [PMID: 32751422 PMCID: PMC7432006 DOI: 10.3390/ijerph17155493] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/22/2020] [Accepted: 07/25/2020] [Indexed: 12/17/2022]
Abstract
The primary goal of this pilot study was to assess feasibility of studies among local community members to address the hypothesis that complex exposures to the World Trade Center (WTC) dust and fumes resulted in long-term epigenetic changes. We enrolled 18 WTC-exposed cancer-free women from the WTC Environmental Health Center (WTC EHC) who agreed to donate blood samples during their standard clinical visits. As a reference WTC unexposed group, we randomly selected 24 age-matched cancer-free women from an existing prospective cohort who donated blood samples before 11 September 2001. The global DNA methylation analyses were performed using Illumina Infinium MethylationEpic arrays. Statistical analyses were performed using R Bioconductor package. Functional genomic analyses were done by mapping the top 5000 differentially expressed CpG sites to the Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway database. Among cancer-free subjects, we observed substantial methylation differences between WTC-exposed and unexposed women. The top 15 differentially methylated gene probes included BCAS2, OSGIN1, BMI1, EEF1A2, SPTBN5, CHD8, CDCA7L, AIDA, DDN, SNORD45C, ZFAND6, ARHGEF7, UBXN8, USF1, and USP12. Several cancer-related pathways were enriched in the WTC-exposed subjects, including endocytosis, mitogen-activated protein kinase (MAPK), viral carcinogenesis, as well as Ras-associated protein-1 (Rap1) and mammalian target of rapamycin (mTOR) signaling. The study provides preliminary data on substantial differences in DNA methylation between WTC-exposed and unexposed populations that require validation in further studies.
Collapse
Affiliation(s)
- Alan A. Arslan
- Department of Obstetrics and Gynecology, New York University Langone Health, New York, NY 10016, USA
- Department of Population Health, New York University Langone Health, New York, NY 10016, USA; (S.T.); (L.Y.); (Y.Z.); (A.Z.-J.); (Y.S.)
- NYU Perlmutter Comprehensive Cancer Center, New York, NY 10016, USA
- Correspondence:
| | - Stephanie Tuminello
- Department of Population Health, New York University Langone Health, New York, NY 10016, USA; (S.T.); (L.Y.); (Y.Z.); (A.Z.-J.); (Y.S.)
| | - Lei Yang
- Department of Population Health, New York University Langone Health, New York, NY 10016, USA; (S.T.); (L.Y.); (Y.Z.); (A.Z.-J.); (Y.S.)
| | - Yian Zhang
- Department of Population Health, New York University Langone Health, New York, NY 10016, USA; (S.T.); (L.Y.); (Y.Z.); (A.Z.-J.); (Y.S.)
| | - Nedim Durmus
- Department of Medicine, New York University Langone Health, New York, NY 10016, USA; (N.D.); (J.R.)
| | - Matija Snuderl
- Department of Pathology, New York University Langone Health, New York, NY 10016, USA; (M.S.); (A.H.)
| | - Adriana Heguy
- Department of Pathology, New York University Langone Health, New York, NY 10016, USA; (M.S.); (A.H.)
- NYU Langone’s Genome Technology Center, New York, NY 10016, USA
| | - Anne Zeleniuch-Jacquotte
- Department of Population Health, New York University Langone Health, New York, NY 10016, USA; (S.T.); (L.Y.); (Y.Z.); (A.Z.-J.); (Y.S.)
- NYU Perlmutter Comprehensive Cancer Center, New York, NY 10016, USA
| | - Yongzhao Shao
- Department of Population Health, New York University Langone Health, New York, NY 10016, USA; (S.T.); (L.Y.); (Y.Z.); (A.Z.-J.); (Y.S.)
- NYU Perlmutter Comprehensive Cancer Center, New York, NY 10016, USA
| | - Joan Reibman
- Department of Medicine, New York University Langone Health, New York, NY 10016, USA; (N.D.); (J.R.)
| |
Collapse
|