1
|
Zheng J, Xu X, Zhang Z, Ge K, Xiang Y, Dai H. Magea13 attenuates myocardial injury in acute myocardial infarction by inhibiting the cAMP-PKA signaling pathway. Apoptosis 2025; 30:1042-1057. [PMID: 40056358 PMCID: PMC11946976 DOI: 10.1007/s10495-025-02078-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2025] [Indexed: 03/10/2025]
Abstract
OBJECT Acute myocardial infarction (AMI) is a serious cardiovascular disease for which there are still no effective therapeutic options available, and melanoma-associated antigen-A13 (Magea13), a member of the MAGE superfamily, has an unknown role in AMI. This study aims to investigate the potential role and molecular mechanisms of Magea13 in myocardial injury associated with AMI through in vivo and in vitro experiments. METHODS Firstly, differentially expressed genes (DEGs) and signaling pathways were screened by RNA sequencing. Cardiac-specific Magea13 overexpression was achieved with the adeno-associated virus type 9 serotype system. Subsequently, these rats underwent left anterior descending coronary artery (LAD) ligation, followed by histopathological examination, biochemical assay, and Western blot analysis to evaluate the efficacy and feasibility of Magea13 in AMI. Meanwhile, the Magea13-overexpressing rat cardiomyocyte cell line (H9c2) was also subjected to hypoxia-glucose deficiency/reperfusion to mimic AMI injury to further validate its effects in vitro. RESULTS The cardiomyocyte-specific overexpression of Magea13 was observed to attenuate myocardial injury in rats with acute myocardial infarction. Furthermore, Magea13 overexpression was demonstrated to attenuate OGD/R-induced H9c2 cell injury. Mechanistic studies have suggested that the protective effect of Magea13 may be mediated through the cAMP-PKA pathway. CONCLUSION Magea13 has been demonstrated to offer protection against AMI myocardial injury through the cAMP-PKA signaling pathway and is therefore a promising therapeutic and predictive target for AMI myocardial injury.
Collapse
Affiliation(s)
- Jialin Zheng
- Department of Cardiology, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming city, Yunnan Province, 650102, China
| | - Xiaoyu Xu
- Department of Cardiology, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming city, Yunnan Province, 650102, China
| | - Ziwei Zhang
- Department of Cardiology, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming city, Yunnan Province, 650102, China
| | - Kanghui Ge
- Department of Cardiology, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming city, Yunnan Province, 650102, China
| | - Yi Xiang
- Department of Cardiology, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming city, Yunnan Province, 650102, China
| | - Hualei Dai
- Department of Cardiology, The Second People's Hospital of Yunnan Province/Affiliated Hospital of Yunnan University, No.176 Qingnian Road, Wuhua District, Kunming city, Yunnan Province, 650021, China.
| |
Collapse
|
2
|
Mohsenzadegan M, Fattahi F, Kalantari E, Abolhasani M, Madjd Z, Saeednejad Zanjani L. Overexpression of MAGE-A2 is Related to the Malignant Degree and Progression of Disease in Patients With Clear Cell Renal Cell Carcinoma. Appl Immunohistochem Mol Morphol 2025; 33:78-90. [PMID: 39801191 DOI: 10.1097/pai.0000000000001243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 12/17/2024] [Indexed: 03/06/2025]
Abstract
Melanoma antigen gene-A2 (MAGE-A2) is one of the most cancer-testis antigens overexpressed in a variety of malignancies. However, the expression of MAGE-A2 for clinical values in the pathophysiology of renal cell carcinoma (RCC) is unknown. For the first time, the present study was conducted to examine the expression and prognostic significance of MAGE-A2 expression in clear cell RCC (ccRCC). MAGE-A2 expression was assayed in 162 well-defined ccRCC samples using immunohistochemistry staining on tissue microarrays. The association between MAGE-A2 expression and clinic-pathologic features as well as survival outcomes were then performed. A significant and positive correlation was found between cytoplasmic expression of MAGE-A2 with tumor size ( P =0.008), nucleolar grade ( P =0.001), tumor stage ( P =0.001), microvascular invasion ( P =0.001), invasion to renal pelvis ( P =0.032), renal sinus fat ( P =0.004), and Gerota's fascia ( P =0.028) as well as histologic tumor necrosis ( P <0.0001). Increased expression of MAGE-A2 was observed to be associated with shorter progression-free survival (PFS) compared with patients with low expression of MAGE-A2 ( P =0.032). Multivariate analysis revealed that tumor size and nucleolar grade are independent predictors of the PFS ( P =0.054, P =0.032, respectively). Our results indicated that increased cytoplasmic expression of MAGE-A2 is associated with the malignant degree and progression of ccRCC. This data improved the significance of MAGE-A2 expression and will potentially allow using MAGE-A2 for the prognosis of the disease and immunotherapy in patients with ccRCC.
Collapse
Affiliation(s)
| | - Fahimeh Fattahi
- Oncopathology Research Center
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
| | | | - Maryam Abolhasani
- Oncopathology Research Center
- Hasheminejad Kidney Center, Iran University of Medical Sciences, (IUMS), Tehran
| | | | - Leili Saeednejad Zanjani
- Oncopathology Research Center
- Department of Pathology and Genomic Medicine, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
3
|
Sabatelle RC, Colson YL, Sachdeva U, Grinstaff MW. Drug Delivery Opportunities in Esophageal Cancer: Current Treatments and Future Prospects. Mol Pharm 2024; 21:3103-3120. [PMID: 38888089 PMCID: PMC11331583 DOI: 10.1021/acs.molpharmaceut.4c00246] [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] [Indexed: 06/20/2024]
Abstract
With one of the highest mortality rates of all malignancies, the 5-year survival rate for esophageal cancer is under 20%. Depending on the stage and extent of the disease, the current standard of care treatment paradigm includes chemotherapy or chemoradiotherapy followed by surgical esophagogastrectomy, with consideration for adjuvant immunotherapy for residual disease. This regimen has high morbidity, due to anatomic changes inherent in surgery, the acuity of surgical complications, and off-target effects of systemic chemotherapy and immunotherapy. We begin with a review of current treatments, then discuss new and emerging targets for therapies and advanced drug delivery systems. Recent and ongoing preclinical and early clinical studies are evaluating traditional tumor targets (e.g., human epidermal growth factor receptor 2), as well as promising new targets such as Yes-associated protein 1 or mammalian target of rapamycin to develop new treatments for this disease. Due the function and location of the esophagus, opportunities also exist to pair these treatments with a drug delivery strategy to increase tumor targeting, bioavailability, and intratumor concentrations, with the two most common delivery platforms being stents and nanoparticles. Finally, early results with antibody drug conjugates and chimeric antigenic receptor T cells show promise as upcoming therapies. This review discusses these innovations in therapeutics and drug delivery in the context of their successes and failures, with the goal of identifying those solutions that demonstrate the most promise to shift the paradigm in treating this deadly disease.
Collapse
Affiliation(s)
- Robert C. Sabatelle
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA, 02215, USA
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Uma Sachdeva
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Mark W. Grinstaff
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA, 02215, USA
| |
Collapse
|
4
|
Wang ZW, Yu QY, Xu MJ, Zhou CY, Li JP, Liao XH. MAGE-A11 is a potential prognostic biomarker and immunotherapeutic target in gastric cancer. Aging (Albany NY) 2024; 16:285-298. [PMID: 38180746 PMCID: PMC10817374 DOI: 10.18632/aging.205368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/15/2023] [Indexed: 01/06/2024]
Abstract
Gastric cancer poses a serious threat to human health and affects the digestive system. The lack of early symptoms and a dearth of effective identification methods make diagnosis difficult, with many patients only receiving a definitive diagnosis at a malignant stage, causing them to miss out on optimal therapeutic interventions. Melanoma-associated antigen-A (MAGE-A) is part of the MAGE family and falls under the cancer/testis antigen (CTA) category. The MAGE-A subfamily plays a significant role in tumorigenesis, proliferation and migration. The expression, prognosis and function of MAGE-A family members in GC, however, remain unclear. Our research and screening have shown that MAGE-A11 was highly expressed in GC tissues and was associated with poor patient prognosis. Additionally, MAGE-A11 functioned as an independent prognostic factor in GC through Cox regression analysis, and its expression showed significant correlation with both tumour immune cell infiltration and responsiveness to immunotherapy. Our data further indicated that MAGE-A11 regulated GC cell proliferation and migration. Subsequently, our findings propose that MAGE-A11 may operate as a prognostic factor, having potential as an immunotherapy target for GC.
Collapse
Affiliation(s)
- Zhi-Wen Wang
- Institute of Biology and Medicine, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P.R. China
- Key Laboratory of Chronic Noncommunicable Diseases, Yueyang Vocational Technical College, Yueyang 414006, Hunan, P.R. China
| | - Qi-Ying Yu
- Central Laboratory, Tumor Hospital Affiliated to Nantong University, Nantong 226361, Jiangsu, P.R. China
| | - Meng-Jiao Xu
- Zhaoyuan Linglong Central Health Center, Zhaoyuan 265400, Shandong, P.R. China
| | - Chuan-Yi Zhou
- Yueyang People’s Hospital, Yueyang Hospital Affiliated to Hunan Normal University Neoplasm Ward 1, Yueyang 414000, Hunan, P.R. China
| | - Jia-Peng Li
- Institute of Biology and Medicine, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P.R. China
- College of Science, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P.R. China
| | - Xing-Hua Liao
- Institute of Biology and Medicine, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P.R. China
| |
Collapse
|
5
|
Qin H, Chen J, Bouchekioua-Bouzaghou K, Meng YM, Griera JB, Jiang X, Kong X, Wang M, Xu Q, Wong PP. Immunization with a multi-antigen targeted DNA vaccine eliminates chemoresistant pancreatic cancer by disrupting tumor-stromal cell crosstalk. J Transl Med 2023; 21:702. [PMID: 37814317 PMCID: PMC10561406 DOI: 10.1186/s12967-023-04519-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/11/2023] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is characterised by limited responses to chemoimmunotherapy attributed to highly desmoplastic tumor microenvironment. Disrupting the tumor-stromal cell crosstalk is considered as an improved PDAC treatment strategy, whereas little progress has been made due to poor understanding of its underlying mechanism. Here, we examined the cellular role of melanoma associated antigen A isoforms (MAGEA) in regulating tumor-stromal crosstalk mediated chemoresistance. METHODS We used clinical samples to explore the correlation between MAGEA expression and patient prognosis in multiple cancers. We utilized cancer cell lines, patient derived organoids and orthotopic PDAC model to examine the function of MAGEA in chemoresistance. We performed biochemical, proteome profiler array and transcriptional analysis to uncover a mechanism that governs tumor-stromal crosstalk. We developed a multi-MAGEA antigen targeted DNA vaccine and tested its effect on PDAC tumor growth. RESULTS We establish MAGEA as a regulator of the tumor-stromal crosstalk in PDAC. We provide strong clinical evidence indicating that high MAGEA expression, including MAGEA2, MAGEA3 and MAGEA10, correlates with worse chemotherapeutic response and poor prognosis in multiple cancers, while their expression is up-regulated in chemoresistant PDAC patient derived organoids and cancer cell lines. Mechanistically, MAGEA2 prohibits gemcitabine-induced JNK-c-Jun-p53 mediated cancer cell apoptosis, while gemcitabine stimulated pancreatic stellate cells secretes GDF15 to further enhance the gemcitabine resistance of MAGEA2 expressing cells by activating GFRAL-RET mediated Akt and ERK1/2 dependent survival pathway. Strikingly, immunization with a DNA vaccine that targeting multiple MAGEA antigens, including MAGEA2, MAGEA3 and MAGEA10, elicits robust immune responses against the growth of gemcitabine resistant tumors. CONCLUSIONS These findings suggest that targeting MAGEA-mediated paracrine regulation of chemoresistance by immunotherapy can be an improved pancreatic cancer treatment strategy.
Collapse
Affiliation(s)
- Hongquan Qin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Jiali Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Katia Bouchekioua-Bouzaghou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Ya-Ming Meng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine; Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Reserach Center for Obstetrics and Gynecology; Guangdong-HongKong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jordi Bach Griera
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Xue Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Xiangzhan Kong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Minghui Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Qiuping Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Ping-Pui Wong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| |
Collapse
|
6
|
Fabrication of poly (aspartic) acid functionalized graphene quantum dots based FRET sensor for selective and sensitive detection of MAGE-A11 antigen. Microchem J 2022. [DOI: 10.1016/j.microc.2022.107971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
7
|
Liu M, Li J, Wang Y, Ghaffar M, Yang Y, Wang M, Li C. MAGEA6 positively regulates MSMO1 and promotes the migration and invasion of oesophageal cancer cells. Exp Ther Med 2022; 23:204. [PMID: 35126707 PMCID: PMC8796618 DOI: 10.3892/etm.2022.11127] [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: 11/17/2020] [Accepted: 08/02/2021] [Indexed: 11/20/2022] Open
Abstract
The melanoma antigen gene family A (MAGEA) family of proteins comprises of cancer-testis antigens that are highly expressed in a number of tumours but are minimally expressed in normal cells. Due to its expression characteristics, this protein family has become a popular target for anti-cancer drugs and immunotherapy research over recent years. Although, elevated expression levels of MAGEA6 has been found in different types of tumours, there remains to be insufficient information on the function of MAGEA6 and its associated gene regulation pathways. The present study used Transwell, Cell Counting Kit-8 and wound healing assays to analyse the effects of MAGEA6 on Eca109 cell invasion, migration and proliferation. The main functions and pathways involved in MAGEA6 were predicted by Illumina Hiseq screening for mutually regulated genes and core genes. Eca109 cell line with a high expression of MAGEA6 was a stable cell line obtained by transfection in the early stage, and this cell line was used in subsequent experiments. Transcriptome sequencing was performed on this cell line and the Eca109 cell line that normally expressed MAGEA6. It was revealed that a high expression of MAGEA6 conferred a significant stimulating effect on cell proliferation whilst also significantly increasing cell invasion and migration. Transcriptomic analysis identified 14 differentially expressed genes and 13 core regulatory genes closely associated with MAGEA6 expression regulation, such as methylsterol monooxygenase 1 (MSMO1). The present study suggest that MAGEA6 positively regulated MSMO1 expression, which may serve an oncogenic role in cells through this regulatory effect. Overall, this provided a novel route of investigation for an in-depth study of the regulatory function of MAGEA6.
Collapse
Affiliation(s)
- Manyu Liu
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Jintao Li
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Yangjunqi Wang
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Maliha Ghaffar
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Yishu Yang
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Minglian Wang
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| | - Changshuo Li
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, P.R. China
| |
Collapse
|
8
|
MAGEA4 Coated Extracellular Vesicles Are Stable and Can Be Assembled In Vitro. Int J Mol Sci 2021; 22:ijms22105208. [PMID: 34069064 PMCID: PMC8155938 DOI: 10.3390/ijms22105208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/02/2021] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) are valued candidates for the development of new tools for medical applications. Vesicles carrying melanoma-associated antigen A (MAGEA) proteins, a subfamily of cancer-testis antigens, are particularly promising tools in the fight against cancer. Here, we have studied the biophysical and chemical properties of MAGEA4-EVs and show that they are stable under common storage conditions such as keeping at +4 °C and -80 °C for at least 3 weeks after purification. The MAGEA4-EVs can be freeze-thawed two times without losing MAGEA4 in detectable quantities. The attachment of MAGEA4 to the surface of EVs cannot be disrupted by high salt concentrations or chelators, but the vesicles are sensitive to high pH. The MAGEA4 protein can bind to the surface of EVs in vitro, using robust passive incubation. In addition, EVs can be loaded with recombinant proteins fused to the MAGEA4 open reading frame within the cells and also in vitro. The high stability of MAGEA4-EVs ensures their potential for the development of EV-based anti-cancer applications.
Collapse
|
9
|
Ladelfa MF, Peche LY, Amato GE, Escalada MC, Zampieri S, Pascucci FA, Benevento AF, Do Porto DF, Dardis A, Schneider C, Monte M. Expression of the tumor-expressed protein MageB2 enhances rRNA transcription. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119015. [PMID: 33741433 DOI: 10.1016/j.bbamcr.2021.119015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/22/2021] [Accepted: 03/12/2021] [Indexed: 11/17/2022]
Abstract
An essential requirement for cells to sustain a high proliferating rate is to be paired with enhanced protein synthesis through the production of ribosomes. For this reason, part of the growth-factor signaling pathways, are devoted to activate ribosome biogenesis. Enhanced production of ribosomes is a hallmark in cancer cells, which is boosted by different mechanisms. Here we report that the nucleolar tumor-protein MageB2, whose expression is associated with cell proliferation, also participates in ribosome biogenesis. Studies carried out in both siRNA-mediated MageB2 silenced cells and CRISPR/CAS9-mediated MageB2 knockout (KO) cells showed that its expression is linked to rRNA transcription increase independently of the cell proliferation status. Mechanistically, MageB2 interacts with phospho-UBF, a protein which causes the recruitment of RNA Pol I pre-initiation complex required for rRNA transcription. In addition, cells expressing MageB2 displays enhanced phospho-UBF occupancy at the rDNA gene promoter. Proteomic studies performed in MageB2 KO cells revealed impairment in ribosomal protein (RPs) content. Functionally, enhancement in rRNA production in MageB2 expressing cells, was directly associated with an increased dynamic in protein synthesis. Altogether our results unveil a novel function for a tumor-expressed protein from the MAGE-I family. Findings reported here suggest that nucleolar MageB2 might play a role in enhancing ribosome biogenesis as part of its repertoire to support cancer cell proliferation.
Collapse
Affiliation(s)
- María Fátima Ladelfa
- Lab. Oncología Molecular, Departamento de Química Biológica, IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Leticia Yamila Peche
- Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, Area Science Park, Trieste, Italy
| | - Gastón Ezequiel Amato
- Lab. Oncología Molecular, Departamento de Química Biológica, IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Micaela Carolina Escalada
- Lab. Oncología Molecular, Departamento de Química Biológica, IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Stefania Zampieri
- Centro di Coordinamento Regionale per le Malattie Rare, Ospedale Universitario Santa Maria Della Misericordia, Udine, Italy
| | - Franco Andrés Pascucci
- Lab. Oncología Molecular, Departamento de Química Biológica, IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Andres Fernandez Benevento
- Plataforma de Bioinformática Argentina, Instituto de Cálculo, Pabellón 2, Ciudad Universitaria, Facultad de Ciencias Exactas y Naturales, UBA, Buenos Aires, Argentina
| | - Dario Fernandez Do Porto
- Plataforma de Bioinformática Argentina, Instituto de Cálculo, Pabellón 2, Ciudad Universitaria, Facultad de Ciencias Exactas y Naturales, UBA, Buenos Aires, Argentina
| | - Andrea Dardis
- Centro di Coordinamento Regionale per le Malattie Rare, Ospedale Universitario Santa Maria Della Misericordia, Udine, Italy
| | - Claudio Schneider
- Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, Area Science Park, Trieste, Italy; Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, p.le Kolbe 4, Udine, Italy
| | - Martin Monte
- Lab. Oncología Molecular, Departamento de Química Biológica, IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
10
|
Zhu M, Wang S. Functional Nucleic‐Acid‐Decorated Spherical Nanoparticles: Preparation Strategies and Current Applications in Cancer Therapy. SMALL SCIENCE 2021. [DOI: 10.1002/smsc.202000056] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Min Zhu
- Department of Pharmaceutical Engineering College of Chemistry and Chemical Engineering Central South University No. 932 South Lushan Rd Changsha Hunan 410083 P. R. China
| | - Shan Wang
- Department of Pharmaceutical Engineering College of Chemistry and Chemical Engineering Central South University No. 932 South Lushan Rd Changsha Hunan 410083 P. R. China
| |
Collapse
|
11
|
Vacchelli E, Martins I, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Peptide vaccines in cancer therapy. Oncoimmunology 2021; 1:1557-1576. [PMID: 23264902 PMCID: PMC3525611 DOI: 10.4161/onci.22428] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Prophylactic vaccination constitutes one of the most prominent medical achievements of history. This concept was first demonstrated by the pioneer work of Edward Jenner, dating back to the late 1790s, after which an array of preparations that confer life-long protective immunity against several infectious agents has been developed. The ensuing implementation of nation-wide vaccination programs has de facto abated the incidence of dreadful diseases including rabies, typhoid, cholera and many others. Among all, the most impressive result of vaccination campaigns is surely represented by the eradication of natural smallpox infection, which was definitively certified by the WHO in 1980. The idea of employing vaccines as anticancer interventions was first theorized in the 1890s by Paul Ehrlich and William Coley. However, it soon became clear that while vaccination could be efficiently employed as a preventive measure against infectious agents, anticancer vaccines would have to (1) operate as therapeutic, rather than preventive, interventions (at least in the vast majority of settings), and (2) circumvent the fact that tumor cells often fail to elicit immune responses. During the past 30 y, along with the recognition that the immune system is not irresponsive to tumors (as it was initially thought) and that malignant cells express tumor-associated antigens whereby they can be discriminated from normal cells, considerable efforts have been dedicated to the development of anticancer vaccines. Some of these approaches, encompassing cell-based, DNA-based and purified component-based preparations, have already been shown to exert conspicuous anticancer effects in cohorts of patients affected by both hematological and solid malignancies. In this Trial Watch, we will summarize the results of recent clinical trials that have evaluated/are evaluating purified peptides or full-length proteins as therapeutic interventions against cancer.
Collapse
Affiliation(s)
- Erika Vacchelli
- Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM, U848; Villejuif, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Wang CY, Lin BL, Chen CH. Targeted drug delivery using an aptamer against shared tumor-specific peptide antigen of MAGE-A3. Cancer Biol Ther 2020; 22:12-18. [PMID: 33249980 DOI: 10.1080/15384047.2020.1833156] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
We developed a DNA aptamer, Ap52, against the shared tumor-specific MAGE-A3111-125 peptide antigen that was used to target multiple types of cancer cells. Here we report the in vivo study of mice implanted with pancreatic tumor cells AsPC-1, which demonstrates accumulation of phosphorothioate-modified Ap52 (ThioAp52) at the xenograft tumor following either intravenous or in situ injection. When complexed with antitumor drug doxorubicin (Dox), ThioAp52 achieves targeted delivery to four types of cancer cells, including breast, oral, pancreatic, and skin. Image analysis shows that ThioAp52-Dox complex selectively enters cancer cells, while free Dox is taken up by all cell lines. The cytotoxicity of ThioAp52-Dox for cancer cells is enhanced as compared to that for the corresponding normal/noncancerous cells. These results indicate that this aptamer against shared tumor-specific antigen can be a potential delivery vehicle for therapeutics to treat multiple cancers.
Collapse
Affiliation(s)
- Chin-Yu Wang
- Genomics Research Center, Academia Sinica , Taipei, Taiwan
| | - Bai-Ling Lin
- Genomics Research Center, Academia Sinica , Taipei, Taiwan
| | | |
Collapse
|
13
|
Liu S, Zhao Y, Xu Y, Sang M, Zhao R, Gu L, Shan B. MAGE-A genes as predictors of the outcome of laryngeal squamous cell carcinoma. Oncol Lett 2020; 20:59. [PMID: 32793312 DOI: 10.3892/ol.2020.11920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 06/29/2020] [Indexed: 01/02/2023] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is one of the most common malignant tumors in the head and neck area. Melanoma-associated antigens A (MAGE-A) are strictly tumor-specific and are expressed in several types of tumors. To date, no studies have reported the potential of MAGE-A genes as markers for circulating tumor cells (CTCs) in patients with LSCC. The present study aimed to evaluate the expression and the possible prognostic significance of MAGE-A in the peripheral blood of patients with LSCC. In the present study, the expression of MAGE-A genes was determined by multiplex semi-nested PCR and restriction endonuclease treatment of the peripheral blood of patients with LSCC. The association between MAGE-A gene expression and clinicopathological parameters and prognosis was evaluated. The results demonstrated that the expression of MAGE-A was associated with the predictors that indicate poor prognosis. The expression levels of MAGE-A and each individual MAGE-A gene were also associated with a shorter overall survival time of patients with LSCC. In conclusion, the results of the present study suggested that the expression of MAGE-A genes may be a potential prognostic marker for patients with LSCC.
Collapse
Affiliation(s)
- Shenghui Liu
- Department of Otolaryngology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Yan Zhao
- Department of Otolaryngology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Yuru Xu
- Department of Otolaryngology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Meixiang Sang
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Ruili Zhao
- Department of Otolaryngology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Lina Gu
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Baoen Shan
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|
14
|
Niemira M, Collin F, Szalkowska A, Bielska A, Chwialkowska K, Reszec J, Niklinski J, Kwasniewski M, Kretowski A. Molecular Signature of Subtypes of Non-Small-Cell Lung Cancer by Large-Scale Transcriptional Profiling: Identification of Key Modules and Genes by Weighted Gene Co-Expression Network Analysis (WGCNA). Cancers (Basel) 2019; 12:E37. [PMID: 31877723 PMCID: PMC7017323 DOI: 10.3390/cancers12010037] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) represents a heterogeneous group of malignancies consisting essentially of adenocarcinoma (ADC) and squamous cell carcinoma (SCC). Although the diagnosis and treatment of ADC and SCC have been greatly improved in recent decades, there is still an urgent need to identify accurate transcriptome profile associated with the histological subtypes of NSCLC. The present study aims to identify the key dysregulated pathways and genes involved in the development of lung ADC and SCC and to relate them with the clinical traits. The transcriptional changes between tumour and normal lung tissues were investigated by RNA-seq. Gene ontology (GO), canonical pathways analysis with the prediction of upstream regulators, and weighted gene co-expression network analysis (WGCNA) to identify co-expressed modules and hub genes were used to explore the biological functions of the identified dysregulated genes. It was indicated that specific gene signatures differed significantly between ADC and SCC related to the distinct pathways. Of identified modules, four and two modules were the most related to clinical features in ADC and SCC, respectively. CTLA4, MZB1, NIP7, and BUB1B in ADC, as well as GNG11 and CCNB2 in SCC, are novel top hub genes in modules associated with tumour size, SUVmax, and recurrence-free survival. Our research provides a more effective understanding of the importance of biological pathways and the relationships between major genes in NSCLC in the perspective of searching for new molecular targets.
Collapse
Affiliation(s)
- Magdalena Niemira
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (A.S.); (A.B.); (A.K.)
| | - Francois Collin
- Centre for Bioinformatics and Data Analysis, Medical University of Bialystok, 15-276 Bialystok, Poland; (F.C.); (K.C.); (M.K.)
| | - Anna Szalkowska
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (A.S.); (A.B.); (A.K.)
| | - Agnieszka Bielska
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (A.S.); (A.B.); (A.K.)
| | - Karolina Chwialkowska
- Centre for Bioinformatics and Data Analysis, Medical University of Bialystok, 15-276 Bialystok, Poland; (F.C.); (K.C.); (M.K.)
| | - Joanna Reszec
- Department of Medical Pathomorphology, Medical University of Bialystok, 15-276 Bialystok, Poland;
| | - Jacek Niklinski
- Department of Clinical Molecular Biology, Medical University of Bialystok, 15-276 Bialystok, Poland;
| | - Miroslaw Kwasniewski
- Centre for Bioinformatics and Data Analysis, Medical University of Bialystok, 15-276 Bialystok, Poland; (F.C.); (K.C.); (M.K.)
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (A.S.); (A.B.); (A.K.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| |
Collapse
|
15
|
Sanderson JP, Crowley DJ, Wiedermann GE, Quinn LL, Crossland KL, Tunbridge HM, Cornforth TV, Barnes CS, Ahmed T, Howe K, Saini M, Abbott RJ, Anderson VE, Tavano B, Maroto M, Gerry AB. Preclinical evaluation of an affinity-enhanced MAGE-A4-specific T-cell receptor for adoptive T-cell therapy. Oncoimmunology 2019; 9:1682381. [PMID: 32002290 PMCID: PMC6959444 DOI: 10.1080/2162402x.2019.1682381] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/12/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022] Open
Abstract
A substantial obstacle to the success of adoptive T cell-based cancer immunotherapy is the sub-optimal affinity of T-cell receptors (TCRs) for most tumor antigens. Genetically engineered TCRs that have enhanced affinity for specific tumor peptide-MHC complexes may overcome this barrier. However, this enhancement risks increasing weak TCR cross-reactivity to other antigens expressed by normal tissues, potentially leading to clinical toxicities. To reduce the risk of such adverse clinical outcomes, we have developed an extensive preclinical testing strategy, involving potency testing using 2D and 3D human cell cultures and primary tumor material, and safety testing using human primary cell and cell-line cross-reactivity screening and molecular analysis to predict peptides recognized by the affinity-enhanced TCR. Here, we describe this strategy using a developmental T-cell therapy, ADP-A2M4, which recognizes the HLA-A2-restricted MAGE-A4 peptide GVYDGREHTV. ADP-A2M4 demonstrated potent anti-tumor activity in the absence of major off-target cross-reactivity against a range of human primary cells and cell lines. Identification and characterization of peptides recognized by the affinity-enhanced TCR also revealed no cross-reactivity. These studies demonstrated that this TCR is highly potent and without major safety concerns, and as a result, this TCR is now being investigated in two clinical trials (NCT03132922, NCT04044768).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Tina Ahmed
- Preclinical Research, Adaptimmune, Abingdon, UK
| | - Karen Howe
- Target Validation, Adaptimmune, Abingdon, UK
| | - Manoj Saini
- Preclinical Research, Adaptimmune, Abingdon, UK
| | | | | | | | | | | |
Collapse
|
16
|
Mao Y, Fan W, Hu H, Zhang L, Michel J, Wu Y, Wang J, Jia L, Tang X, Xu L, Chen Y, Zhu J, Feng Z, Xu L, Yin R, Tang Q. MAGE-A1 in lung adenocarcinoma as a promising target of chimeric antigen receptor T cells. J Hematol Oncol 2019; 12:106. [PMID: 31640756 PMCID: PMC6805483 DOI: 10.1186/s13045-019-0793-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cancer/testis antigens (CTAs) are a special type of tumor antigen and are believed to act as potential targets for cancer immunotherapy. METHODS In this study, we first screened a rational CTA MAGE-A1 for lung adenocarcinoma (LUAD) and explored the detailed characteristics of MAGE-A1 in LUAD development through a series of phenotypic experiments. Then, we developed a novel MAGE-A1-CAR-T cell (mCART) using lentiviral vector based on our previous MAGE-A1-scFv. The anti-tumor effects of this mCART were finally investigated in vitro and in vivo. RESULTS The results showed striking malignant behaviors of MAGE-A1 in LUAD development, which further validated the rationality of MAGE-A1 as an appropriate target for LUAD treatment. Then, the innovative mCART was successfully constructed, and mCART displayed encouraging tumor-inhibitory efficacy in LUAD cells and xenografts. CONCLUSIONS Taken together, our data suggest that MAGE-A1 is a promising candidate marker for LUAD therapy and the MAGE-A1-specific CAR-T cell immunotherapy may be an effective strategy for the treatment of MAGE-A1-positive LUAD.
Collapse
Affiliation(s)
- Yuan Mao
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, The Fourth Clinical College of Nanjing Medical University, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
- NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- Department of Hematology and Oncology, Department of Geriatric Lung Cancer Laboratory, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Weifei Fan
- Department of Hematology and Oncology, Department of Geriatric Lung Cancer Laboratory, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Hao Hu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Louqian Zhang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, The Fourth Clinical College of Nanjing Medical University, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Jerod Michel
- Department of Mathematics, Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Yaqin Wu
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Wang
- Department of Hematology and Oncology, Department of Geriatric Lung Cancer Laboratory, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Lizhou Jia
- NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Xiaojun Tang
- NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Li Xu
- Department of Pathology, Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Chen
- Department of Pathology, Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Zhu
- Huadong Medical Institute of Biotechniques, Nanjing, China
| | - Zhenqing Feng
- NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Lin Xu
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, The Fourth Clinical College of Nanjing Medical University, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China.
| | - Rong Yin
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, The Fourth Clinical College of Nanjing Medical University, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China.
| | - Qi Tang
- NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
17
|
Meng L, Zhou Y, Ju S, Han J, Song C, Kong J, Wu Y, Lu S, Xu J, Yuan W, Zhang E, Wang C, Hu Z, Gu Y, Luo R, Wang X. A cis-eQTL genetic variant in PLK4 confers high risk of hepatocellular carcinoma. Cancer Med 2019; 8:6476-6484. [PMID: 31489978 PMCID: PMC6797585 DOI: 10.1002/cam4.2487] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/23/2019] [Accepted: 07/30/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose The overexpression and knockdown of PLK4 were both reported to generate aneuploidy. Thus, we aimed to investigate whether genetic variants in PLK4 contribute to the development of hepatocellular carcinoma (HCC). Methods We evaluated associations of common variants in PLK4 and its promoter for the risk of HCC in our association study (1300 cases and 1344 controls). The genotype‐tissue expression (GTEx) and The cancer genome atlas (TCGA) databases were used to quantify the expression of PLK4. Cell proliferation and migration affected by PLK4 in HCC were assessed in vitro. Drug susceptibility testing (DST) model was used to assess the sensibility of PLK4‐activated HCC to CFI‐400945, a small molecule inhibitor of PLK4. Results Herein, we found a significant association between rs3811741, located in the PLK4 intron, and liver cancer risk (OR = 1.26, P = 9.81 × 10−5). Although PLK4 expressed at lower levels in somatic tissues compared to the testis, the risk allele A of rs3811741 was associated with increased PLK4 expression in liver cancer tissues. Additionally, PLK4 high expression was remarkably associated with shortened survival of HCC (HR = 1.97, P = .001). Furthermore, overexpression of PLK4 promoted, while knockdown of PLK4 suppressed cancer cell proliferation, migration, and invasion. DST model demonstrated that CFI‐400945 can effectively suppress rampant proliferation of HCC with highly expressed PLK4. Conclusion Taken together, our study demonstrated that PLK4 is a susceptibility gene and plays an oncogenic role in HCC. Furthermore, we identified that PLK4 sensitives HCC to CFI‐400945, which may be an ideal therapy target for HCC.
Collapse
Affiliation(s)
- Lijuan Meng
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yan Zhou
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Sihan Ju
- Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Jing Han
- Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China.,Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Ci Song
- Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Jing Kong
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yifei Wu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Shuai Lu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Jiani Xu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Wenwen Yuan
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Erbao Zhang
- Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Cheng Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Bioinformatics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yayun Gu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Rongcheng Luo
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xuehao Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Liver Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Xu J, Zhu C, Yu Y, Wu W, Cao J, Li Z, Dai J, Wang C, Tang Y, Zhu Q, Wang J, Wen W, Xue L, Zhen F, Liu J, Huang C, Zhao F, Zhou Y, He Z, Pan X, Wei H, Zhu Y, He Y, Que J, Luo J, Chen L, Wang W. Systematic cancer-testis gene expression analysis identified CDCA5 as a potential therapeutic target in esophageal squamous cell carcinoma. EBioMedicine 2019; 46:54-65. [PMID: 31324603 PMCID: PMC6710982 DOI: 10.1016/j.ebiom.2019.07.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 06/21/2019] [Accepted: 07/10/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most lethal malignancies with poor prognosis. Cancer-testis genes (CTGs) have been vigorously pursued as targets for cancer immunotherapy, but the expressive patterns and functional roles of CTGs remain unclear in ESCC. METHODS A systematic screening strategy was adopted to screen CTGs in ESCC by integrating multiple public databases and RNA expression microarray data from 119 ESCC subjects. For the newly identified ESCC prognosis-associated CTGs, an independent cohort of 118 patients with ESCC was recruited to validate the relationship via immunohistochemistry. Furthermore, functional assays were performed to determine the underlying mechanisms. FINDINGS 21 genes were recognized as CTGs, in particular, CDCA5 was aberrantly upregulated in ESCC tissues and significantly associated with poor prognosis (HR = 1.85, 95%CI: 1.14-3.01, P = .013). Immunohistochemical staining confirmed that positive CDCA5 expression was associated with advanced TNM staging and a shorter overall survival rate (45.59% vs 28.00% for CDCA5-/+ subjects, P = 1.86 × 10-3). H3K27 acetylation in CDCA5 promoter might lead to the activation of CDCA5 during ESCC tumorigenesis. Functionally, in vitro assay of gain- and loss-of-function of CDCA5 suggested that CDCA5 could promote ESCC cells proliferation, invasion, migration, apoptosis resistance and reduce chemosensitivity to cisplatin. Moreover, in vivo assay showed that silenced CDCA5 could inhibit tumor growth. Mechanistically, CDCA5 knockdown led to an arrest in G2/M phase and changes in the expression of factors that played fundamental roles in the cell cycle pathway. INTERPRETATION CDCA5 contributed to ESCC progression and might serve as an attractive target for ESCC immunotherapy. FUND: This work was supported by the Natural Science Foundation of Jiangsu Province (No. BK20181083 and BK20181496), Jiangsu Top Expert Program in Six Professions (No. WSW-003 and WSW-007), Major Program of Science and Technology Foundation of Jiangsu Province (No. BE2016790 and BE2018746), Jiangsu Medical Young Talent Project (No. QNRC2016566), the Program of Jiangsu Medical Innovation Team (No. CXTDA2017006), Postgraduate Research & Practice Innovation Program of Jiangsu Province (KYCX18_1487) and Jiangsu Province 333 Talents Project (No. BRA2017545).
Collapse
Affiliation(s)
- Jing Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chengxiang Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Weibing Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Cao
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhihua Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cheng Wang
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Tang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Quan Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Wen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Xue
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fuxi Zhen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinyuan Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chenjun Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhou
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhicheng He
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xianglong Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haixing Wei
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yining Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yaozhou He
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Que
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinghua Luo
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Liang Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Wei Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
19
|
Das B, Senapati S. Functional and mechanistic studies reveal MAGEA3 as a pro-survival factor in pancreatic cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:294. [PMID: 31287009 PMCID: PMC6615156 DOI: 10.1186/s13046-019-1272-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/10/2019] [Indexed: 12/17/2022]
Abstract
Background In the era of personalized therapy, functional annotation of less frequent genetic aberrations will be instrumental in adapting effective therapeutic in clinic. Overexpression of Melanoma associated antigen A3 (MAGEA3) is reported in certain pancreatic cancer (PCA) patients. The major objective of the current study was to investigate the functional role of MAGEA3 in pancreatic cancer cells (PCCs) growth and survival. Methods Using overexpression (tet-on regulated system and constitutive expression system) and knockdown (by siRNA and shRNA) approach, we dissected the mechanistic role of MAGEA3 in pancreatic cancer pathogenesis. We generated MAGEA3 expressing stable PCA cell lines and mouse primary pancreatic epithelial cells. MAGEA3 was also depleted in certain MAGEA3 positive PCCs by siRNA or shRNA. The stable cells were subjected to in vitro assays like proliferation and survival assays under growth factor deprivation or in the presence of cytotoxic drugs. The MAGEA3 overexpressing or depleted stable PCCs were evaluated in vivo using xenograft model to check the role of MAGEA3 in tumor progression. We also dissected the mechanism behind the MAGEA3 role in tumor progression using western blot analysis and CCL2 neutralization. Results MAGEA3 overexpression in PCA cells did not alter the cell proliferation but protected the cells during growth factor deprivation and also in the presence of cytotoxic drugs. However, depletion of MAGEA3 in MAGEA3 positive cells resulted in reduced cell proliferation and increased apoptosis upon growth factor deprivation and also in response to cytotoxic drugs. The in vivo xenograft study revealed that overexpression of MAGEA3 promoted tumor growth however depleting the same hindered the tumor progression. Mechanistically, our in vitro and in vivo study revealed that MAGEA3 has tumor-promoting role by reducing macro-autophagy and overexpressing pro-survival molecules like CCL2 and survivin. Conclusion Our data proves tumor-promoting role of MAGEA3 and provides the rationale to target MAGEA3 and/or its functional mediators like CCL2 for PCA, which may have a better impact in PCA therapy. Electronic supplementary material The online version of this article (10.1186/s13046-019-1272-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Biswajit Das
- Tumor Microenvironment and Animal Models Lab, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India.,Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shantibhusan Senapati
- Tumor Microenvironment and Animal Models Lab, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India.
| |
Collapse
|
20
|
Zhang Y, Zhang Y, Zhang L. Expression of cancer-testis antigens in esophageal cancer and their progress in immunotherapy. J Cancer Res Clin Oncol 2019; 145:281-291. [PMID: 30656409 PMCID: PMC6373256 DOI: 10.1007/s00432-019-02840-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/03/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE Esophageal cancer is a common disease in China with low survival rate due to no obvious early symptoms and lack of effective screening strategies. Traditional treatments usually do not produce desirable results in patients with advanced esophageal cancer, so immunotherapy which relies on tumor-related antigens is needed to combat low survival rates effectively. Cancer-testis antigens (CTA), a large family of tumor-related antigens, have a strong in vivo immunogenicity and tumor-restricted expressing patterns in normal adult tissues. These two characteristics are ideal features of anticancer immunotherapy targets and, therefore, promoted the development of some studies of CTA-based therapy. To provide ideas for the role of the cancer-testis antigens MAGE-A, NY-ESO-1, LAGE-1, and TTK in esophageal cancer, we summarized their expression, prognostic value, and development in immunotherapy. METHODS The relevant literature from PubMed is reviewed in this study. RESULTS In esophageal cancer, although the relationship between expression of MAGE-A, NY-ESO-1, LAGE-1, and TTK and prognosis value is still in a controversial situation, MAGE-A, NY-ESO-1, LAGE-1, and TTK are highly expressed and can induce specific CTL cells to produce particular killing effect on tumor cells, and some clinical trials have demonstrated that immunotherapy for esophageal cancer patients is effective and safe, which provides a new therapeutic strategy for the treatment of esophageal cancer in the future. CONCLUSION In this review, we summarize expression and prognostic value of MAGE-A, NY-ESO-1, LAGE-1, and TTK in esophageal cancer and point out recent advances in immunotherapy about them.
Collapse
Affiliation(s)
- Yujie Zhang
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Yuxin Zhang
- Hepatic Surgery Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Li Zhang
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| |
Collapse
|
21
|
Gao Y, Kardos J, Yang Y, Tamir TY, Mutter-Rottmayer E, Weissman B, Major MB, Kim WY, Vaziri C. The Cancer/Testes (CT) Antigen HORMAD1 promotes Homologous Recombinational DNA Repair and Radioresistance in Lung adenocarcinoma cells. Sci Rep 2018; 8:15304. [PMID: 30333500 PMCID: PMC6192992 DOI: 10.1038/s41598-018-33601-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/01/2018] [Indexed: 12/24/2022] Open
Abstract
The Cancer/Testes (CT) Antigen HORMAD1 is germ cell-restricted and plays developmental roles in generation and processing of meiotic DNA Double Strand Breaks (DSB). Many tumors aberrantly overexpress HORMAD1 yet the potential impact of this CT antigen on cancer biology is unclear. We tested a potential role of HORMAD1 in genome maintenance in lung adenocarcinoma cells. We show that HORMAD1 re-distributes to nuclear foci and co-localizes with the DSB marker γH2AX in response to ionizing radiation (IR) and chemotherapeutic agents. The HORMA domain and C-term disordered oligomerization motif are necessary for localization of HORMAD1 to IR-induced foci (IRIF). HORMAD1-depleted cells are sensitive to IR and camptothecin. In reporter assays, Homologous Recombination (HR)-mediated repair of targeted ISce1-induced DSBs is attenuated in HORMAD1-depleted cells. In Non-Homologous End Joining (NHEJ) reporter assays, HORMAD1-depletion does not affect repair of ISce1-induced DSB. Early DSB signaling events (including ATM phosphorylation and formation of γH2AX, 53BP1 and NBS1 foci) are intact in HORMAD1-depleted cells. However, generation of RPA-ssDNA foci and redistribution of RAD51 to DSB are compromised in HORMAD1-depleted cells, suggesting that HORMAD1 promotes DSB resection. HORMAD1-mediated HR is a neomorphic activity that is independent of its meiotic partners (including HORMAD2 and CCDC36. Bioinformatic analysis of TCGA data show that similar to known HR pathway genes HORMAD1 is overexpressed in lung adenocarcinomas. Overexpression of HR genes is associated with specific mutational profiles (including copy number variation). Taken together, we identify HORMAD1-dependent DSB repair as a new mechanism of radioresistance and a probable determinant of mutability in lung adenocarcinoma.
Collapse
Affiliation(s)
- Yanzhe Gao
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 614 Brinkhous-Bullitt Building, Chapel Hill, NC, 27599, USA
| | - Jordan Kardos
- Lineberger Comprehensive Cancer Center, Curriculum in Genetics and Molecular Biology, and Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Yang Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 614 Brinkhous-Bullitt Building, Chapel Hill, NC, 27599, USA
| | - Tigist Y Tamir
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Elizabeth Mutter-Rottmayer
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 614 Brinkhous-Bullitt Building, Chapel Hill, NC, 27599, USA
| | - Bernard Weissman
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 614 Brinkhous-Bullitt Building, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, Curriculum in Genetics and Molecular Biology, and Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Michael B Major
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - William Y Kim
- Lineberger Comprehensive Cancer Center, Curriculum in Genetics and Molecular Biology, and Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Cyrus Vaziri
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 614 Brinkhous-Bullitt Building, Chapel Hill, NC, 27599, USA. .,Lineberger Comprehensive Cancer Center, Curriculum in Genetics and Molecular Biology, and Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
22
|
Jian W, Li X, Kang J, Lei Y, Bai Y, Xue Y. Antitumor effect of recombinant Mycobacterium smegmatis expressing MAGEA3 and SSX2 fusion proteins. Exp Ther Med 2018; 16:2160-2166. [PMID: 30186454 DOI: 10.3892/etm.2018.6425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 06/08/2018] [Indexed: 12/17/2022] Open
Abstract
Mycobacterium smegmatis (M. smegmatis), which is a nonpathogenic and fast-growing mycobacterium, is a potential vaccine vector capable of expressing heterologous antigens. Spontaneous humoral and cellular immune responses have been demonstrated against cancer/testis antigens (CTA), including melanoma-associated antigen A (MAGEA) and SSX. In the present study, recombinant plasmids expressing MAGEA3 and SSX2 were constructed. The recombinant plasmids were transferred into M. smegmatis to generate the novel antitumor DNA vaccine. As MAGEA3 and SSX2 were in different ligation sequences, the two DNA vaccines were recombinant M. smegmatis MAGEA3-SSX2 (rM.S-MS) and recombinant M. smegmatis SSX2-MAGEA3 (rM.S-SM), respectively. The expression levels of Fusion proteins were assessed by western blotting. BALB/c mice were immunized with rM.S and western blot analysis was used to determine whether antibodies against MAGEA3 or SSX2 were produced in immunized mice. EC9706 cells were inoculated into BALB/c nude mice and the mice were maintained until an obvious visible tumor appeared on the back. Subsequently, the blood from the rM.S immunized BALB/c mice was injected into the BALB/c nude mice via the tail vein. In order to evaluate the antitumor effect of the vaccines, tumor volume and weight were measured 5 to 21 days after injection. Mice were euthanized on day 21 of tumor growth, and the tumor was dissected and weighed. The two fusion proteins were expressed in the rM.S and the specific fusion protein antibodies were expressed in the blood of immunized BALB/c mice. The tumor volumes and weight in the recombinant M. smegmatis MAGEA3 (rM.S-M) and recombinant M. smegmatis SSX2 (rM.S-S) groups were significantly reduced compared with the control group. Furthermore, the decrease in tumor volumes and weight in the rM.S-MS and rM.S-SM groups was more severe than in the rM.S-M or rM.S-S groups. There was no significant difference in the antitumor effect of the rM.S-MS and rM.S-SM groups. The present findings suggest that this rM.S may be a potential candidate therapeutic vaccine for the treatment of cancer.
Collapse
Affiliation(s)
- Wen Jian
- Department of Respiratory Medicine, The First Affiliated Hospital of The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Xin Li
- Medical Oncology Center, Dongguan Kanghua Hospital, Dongguan, Guangdong 523080, P.R. China
| | - Jian Kang
- Department of Basic Microbiology, The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yingfeng Lei
- Department of Basic Microbiology, The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yinlan Bai
- Department of Basic Microbiology, The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Ying Xue
- Medical Oncology Center, Dongguan Kanghua Hospital, Dongguan, Guangdong 523080, P.R. China.,Department of Radiation Oncology, The First Affiliated Hospital of The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|
23
|
Baker AT, Aguirre-Hernández C, Halldén G, Parker AL. Designer Oncolytic Adenovirus: Coming of Age. Cancers (Basel) 2018; 10:E201. [PMID: 29904022 PMCID: PMC6025169 DOI: 10.3390/cancers10060201] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 06/06/2018] [Accepted: 06/11/2018] [Indexed: 12/26/2022] Open
Abstract
The licensing of talimogene laherparepvec (T-Vec) represented a landmark moment for oncolytic virotherapy, since it provided unequivocal evidence for the long-touted potential of genetically modified replicating viruses as anti-cancer agents. Whilst T-Vec is promising as a locally delivered virotherapy, especially in combination with immune-checkpoint inhibitors, the quest continues for a virus capable of specific tumour cell killing via systemic administration. One candidate is oncolytic adenovirus (Ad); it’s double stranded DNA genome is easily manipulated and a wide range of strategies and technologies have been employed to empower the vector with improved pharmacokinetics and tumour targeting ability. As well characterised clinical and experimental agents, we have detailed knowledge of adenoviruses’ mechanisms of pathogenicity, supported by detailed virological studies and in vivo interactions. In this review we highlight the strides made in the engineering of bespoke adenoviral vectors to specifically infect, replicate within, and destroy tumour cells. We discuss how mutations in genes regulating adenoviral replication after cell entry can be used to restrict replication to the tumour, and summarise how detailed knowledge of viral capsid interactions enable rational modification to eliminate native tropisms, and simultaneously promote active uptake by cancerous tissues. We argue that these designer-viruses, exploiting the viruses natural mechanisms and regulated at every level of replication, represent the ideal platforms for local overexpression of therapeutic transgenes such as immunomodulatory agents. Where T-Vec has paved the way, Ad-based vectors now follow. The era of designer oncolytic virotherapies looks decidedly as though it will soon become a reality.
Collapse
Affiliation(s)
- Alexander T Baker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| | - Carmen Aguirre-Hernández
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Gunnel Halldén
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Alan L Parker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| |
Collapse
|
24
|
Õunap K, Kurg K, Võsa L, Maiväli Ü, Teras M, Planken A, Ustav M, Kurg R. Antibody response against cancer-testis antigens MAGEA4 and MAGEA10 in patients with melanoma. Oncol Lett 2018; 16:211-218. [PMID: 29928403 PMCID: PMC6006456 DOI: 10.3892/ol.2018.8684] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 04/19/2018] [Indexed: 02/06/2023] Open
Abstract
Melanoma-associated antigen A (MAGEA) represent a class of tumor antigens that are expressed in a variety of malignant tumors, however, their expression in healthy normal tissues is restricted to germ cells of testis, fetal ovary and placenta. The restricted expression and immunogenicity of these antigens make them ideal targets for immunotherapy in human cancer. In the present study the presence of naturally occurring antibodies against two MAGEA subfamily proteins, MAGEA4 and MAGEA10, was analyzed in patients with melanoma at different stages of disease. Results indicated that the anti-MAGEA4/MAGEA10 immune response in melanoma patients was heterogeneous, with only ~8% of patients having a strong response. Comparing the number of strongly responding patients between different stages of disease revealed that the highest number of strong responses was detected among stage II melanoma patients. These findings support the model that the immune system is involved in the control of melanoma in the early stages of disease.
Collapse
Affiliation(s)
- Kadri Õunap
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia
| | - Kristiina Kurg
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia
| | - Liisi Võsa
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia
| | - Ülo Maiväli
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia
| | - Marina Teras
- Melanoma Unit of The General Surgery and Oncology Surgery Centre, North Estonian Medical Centre, 13419 Tallinn, Estonia
| | - Anu Planken
- Melanoma Unit of The General Surgery and Oncology Surgery Centre, North Estonian Medical Centre, 13419 Tallinn, Estonia
| | - Mart Ustav
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia
| | - Reet Kurg
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia
| |
Collapse
|
25
|
Park S, Sung Y, Jeong J, Choi M, Lee J, Kwon W, Jang S, Park SJ, Kim HS, Lee MH, Kim DJ, Liu K, Kim SH, Dong Z, Ryoo ZY, Kim MO. hMAGEA2 promotes progression of breast cancer by regulating Akt and Erk1/2 pathways. Oncotarget 2018; 8:37115-37127. [PMID: 28415749 PMCID: PMC5514895 DOI: 10.18632/oncotarget.16184] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/06/2017] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most abundant cancer worldwide and a severe problem for women. Notably, breast cancer has a high mortality rate, mainly because of tumor progression and metastasis. Triple-negative breast cancer (TNBC) is highly progressive and lacks the expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Therefore, there are no established therapeutic targets against TNBC. In this study, we investigated whether the expression of human melanoma-associated antigen A2 (MAGEA2) is associated with TNBC. We found that hMAGEA2 is significantly overexpressed in human TNBC tissues; we also observed oncogenic properties using TNBC cell lines (MDA-MB-231 and MDA-MB-468). The overexpression of hMAGEA2 in MDA-MB-231 cell line showed dramatically increased cellular proliferation, colony formation, invasion, and xenograft tumor formation and growth. Conversely, knockdown of hMAEGA2 in MDA-MB-468 cell line suppressed cellular proliferation, colony formation, and xenograft tumor formation. Additionally, we showed that hMAGEA2 regulated the activation of Akt and Erk1/2 signaling pathways. These data indicate that hMAGEA2 is important for progression of TNBC and may serve as a novel molecular therapeutic target.
Collapse
Affiliation(s)
- Song Park
- School of Life Science, BK21 Plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Buk-ku, Daegu, 41566, Republic of Korea
| | - Yonghun Sung
- School of Life Science, BK21 Plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Buk-ku, Daegu, 41566, Republic of Korea
| | - Jain Jeong
- School of Life Science, BK21 Plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Buk-ku, Daegu, 41566, Republic of Korea
| | - Minjee Choi
- School of Life Science, BK21 Plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Buk-ku, Daegu, 41566, Republic of Korea
| | - Jinhee Lee
- School of Life Science, BK21 Plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Buk-ku, Daegu, 41566, Republic of Korea
| | - Wookbong Kwon
- School of Life Science, BK21 Plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Buk-ku, Daegu, 41566, Republic of Korea
| | - Soyoung Jang
- School of Life Science, BK21 Plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Buk-ku, Daegu, 41566, Republic of Korea
| | - Si Jun Park
- School of Life Science, BK21 Plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Buk-ku, Daegu, 41566, Republic of Korea
| | - Hyeng-Soo Kim
- School of Life Science, BK21 Plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Buk-ku, Daegu, 41566, Republic of Korea
| | - Mee-Hyun Lee
- China-US(Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China
| | - Dong Joon Kim
- China-US(Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China
| | - Kangdong Liu
- China-US(Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China
| | - Sung-Hyun Kim
- Institute of Life Science and Biotechnology, Kyungpook National University, Buk-ku, Daegu 41566, Republic of Korea.,China-US(Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China
| | - Zigang Dong
- China-US(Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China
| | - Zae Young Ryoo
- School of Life Science, BK21 Plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Buk-ku, Daegu, 41566, Republic of Korea
| | - Myoung Ok Kim
- The School of Animal BT Science, Kyungpook National University, Sangju-si, Gyeongsangbuk-do 37224, Republic of Korea
| |
Collapse
|
26
|
Kızılbey K, Mansuroğlu B, Derman S, Mustafaeva Akdeste Z. An In vivo study: Adjuvant activity of poly-n-vinyl-2-pyrrolidone-co-acrylic acid on immune responses against Melanoma synthetic peptide. Bioengineered 2018; 9:134-143. [PMID: 28910565 PMCID: PMC5972930 DOI: 10.1080/21655979.2017.1373529] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 07/10/2017] [Accepted: 07/10/2017] [Indexed: 11/23/2022] Open
Abstract
Peptides have been studied as an important class of components in medicine to control many major diseases with vaccination. Polymers as adjuvants are capable of enhancing the vaccine potential against various diseases by improving the delivery of antigens, and they reduce the booster doses of vaccines. In brief, polymers are promising candidates for peptide-based vaccine delivery platforms. The purpose of the present study was to create a possible alternative approach in the treatment of malignant melanoma and/or to prevent metastasis of melanoma. The study was designed as both an experimental and an in vivo study. We prepared a complex and covalent conjugate of MAGE-3 121-134 (L-L-K-Y-R-A-R-E-P-V-T-K-A-E) T-cell epitope as a vaccine candidate for melanoma. These conjugates were able to generate an immune response in mice after a single immunization, without the help of any external adjuvant. The peptide-polymer complexes activated the immune system in the best way and formed the highest antigen specific immune response. These results indicate the adjuvant activity of Poly(N-vinyl-2- pyrrolidone-co-acrylic acid) [P(VP-co-AA)] and the potential use of P(VP-coAA)-peptide based vaccine prototypes for future melanoma cancer vaccine formulations.
Collapse
MESH Headings
- Acrylic Resins/administration & dosage
- Acrylic Resins/chemistry
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/chemistry
- Amino Acid Sequence
- Animals
- Antibodies, Neoplasm/blood
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/chemistry
- Cancer Vaccines/immunology
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/immunology
- Humans
- Immunity, Humoral/drug effects
- Immunization, Secondary
- Immunoconjugates/administration & dosage
- Immunoconjugates/chemistry
- Male
- Melanoma/blood
- Melanoma/immunology
- Melanoma/pathology
- Melanoma/prevention & control
- Mice
- Mice, Inbred BALB C
- Neoplasm Proteins/chemistry
- Neoplasm Proteins/immunology
- Peptides/administration & dosage
- Peptides/chemistry
- Peptides/immunology
- Povidone/administration & dosage
- Povidone/analogs & derivatives
- Povidone/chemistry
- Serum Albumin, Bovine/administration & dosage
- Serum Albumin, Bovine/chemistry
- Serum Albumin, Bovine/immunology
- Skin Neoplasms/blood
- Skin Neoplasms/immunology
- Skin Neoplasms/pathology
- Skin Neoplasms/prevention & control
- T-Lymphocytes/chemistry
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Vaccination
- Vaccines, Subunit
- Melanoma, Cutaneous Malignant
Collapse
Affiliation(s)
- Kadriye Kızılbey
- Biomedical Engineering Department, İstanbul Yeni Yüzyıl University, Yılanlı Ayazma Caddesi, Zeytinburnu-İstanbul, Turkey
| | - Banu Mansuroğlu
- Department of Molecular Biology and Genetics, Yıldız Technical University, Davutpaşa Caddesi, Esenler, İstanbul, Turkey
| | - Serap Derman
- Department of Bioengineering, Yıldız Technical University, Davutpaşa Caddesi, Esenler, İstanbul, Turkey
| | - Zeynep Mustafaeva Akdeste
- Department of Bioengineering, Yıldız Technical University, Davutpaşa Caddesi, Esenler, İstanbul, Turkey
| |
Collapse
|
27
|
Zhou SJ, Wei J, Su S, Chen FJ, Qiu YD, Liu BR. Strategies for Bispecific Single Chain Antibody in Cancer Immunotherapy. J Cancer 2017; 8:3689-3696. [PMID: 29151956 PMCID: PMC5688922 DOI: 10.7150/jca.19501] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 08/07/2017] [Indexed: 12/25/2022] Open
Abstract
Genetic engineering has resulted in more than 50 recombinant bispecific antibody formats over the past two decades. Bispecific scFv antibodies represent a successful and promising immunotherapy platform that retargets cytotoxic T cells to tumor cells, with one scFv directed to tumor-associated antigens and the other to T cells. Based on this antibody construct, strategies for both specific tumor targeting and T cell activation are reviewed here. Three distinct types of tumor antigens are considered to optimize specificity and safety in bispecific scFv based treatment: cancer-testis antigens, neo-antigens and virus-associated antigens. In terms of T cell activation, although CD3 has been widely applied in bispecific scFvs being developed, CD28 and CD137 among co-stimulatory signals are also ideal candidates to be evaluated. Besides, LIGHT and HIV-Tat101 have drawn much attention as their potential roles in modulating antitumor responses.
Collapse
Affiliation(s)
- Shu-Juan Zhou
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Jia Wei
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Shu Su
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Fang-Jun Chen
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Yu-Dong Qiu
- Department of Hepatopancreatobiliary Surgery, The Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, China
| | - Bao-Rui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| |
Collapse
|
28
|
Hossain NM, Chapuis AG, Walter RB. T-Cell Receptor-Engineered Cells for the Treatment of Hematologic Malignancies. Curr Hematol Malig Rep 2017; 11:311-7. [PMID: 27095318 DOI: 10.1007/s11899-016-0327-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Recent attention in adoptive immunotherapy for hematologic malignancies has focused on lymphocytes expressing chimeric antigen receptors. An alternative technique to redirect the immune system toward cancer cells involves the use of T-cells carrying an engineered tumor-recognizing T-cell receptor (TCR). This approach allows targeting of surface or intracellular/nuclear proteins as long as they are processed and presented on the cell surface by human leukocyte antigen molecules. Several trials in advanced solid tumors, particularly melanoma and synovial sarcoma, support the validity of this strategy, although tumor responses have often been short-lived. Emerging data from patients with multiple myeloma and myeloid neoplasms suggest that the benefit of TCR-modified cells may extend to blood cancers. Methodological refinements may be necessary to increase the in vivo persistence and functionality of these cells. Particularly with affinity-enhanced TCRs, however, more effective therapies may increase the potential for serious toxicity due to the unexpected on- or off-target reactivity.
Collapse
Affiliation(s)
- Nasheed M Hossain
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Aude G Chapuis
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, D2-190, Seattle, WA, 98109-1024, USA.,Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, D2-190, Seattle, WA, 98109-1024, USA. .,Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA. .,Department of Epidemiology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
29
|
Park S, Sung Y, Jeong J, Choi M, Lee J, Kwon W, Jang S, Park SJ, Kim JY, Kim SH, Yoon D, Ryoo ZY, Kim MO. Critical roles of hMAGEA2 in induced pluripotent stem cell pluripotency, proliferation, and differentiation. Cell Biochem Funct 2017; 35:392-400. [PMID: 28895148 DOI: 10.1002/cbf.3286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 03/30/2017] [Accepted: 07/07/2017] [Indexed: 12/20/2022]
Abstract
Induced pluripotent stem (iPS) cells are important for clinical application and stem cell research. Although human melanoma-associated antigen A2 (hMAGEA2) expression is known to affect differentiation in embryonic stem cells, its specific role in iPS cells remains unclear. To evaluate the function of hMAGEA2 and its characteristics in iPS cells, we produced hMAGEA2-overexpressing iPS cells from hMAGEA2-overexpressing transgenic mice. Although the iPS cells with overexpressed hMAGEA2 did not differ in morphology, their pluripotency, and self-renewal related genes (Nanog, Oct3/4, Sox2, and Stat3), expression level was significantly upregulated. Moreover, hMAGEA2 contributed to the promotion of cell cycle progression, thereby accelerating cell proliferation. Through embryoid body formation in vitro and teratoma formation in vivo, we demonstrated that hMAGEA2 critically decreases the differentiation ability of iPS cells. These data indicate that hMAGEA2 intensifies the self-renewal, pluripotency, and degree of proliferation of iPS cells, while significantly repressing their differentiation efficiency. Therefore, our findings prove that hMAGEA2 plays key roles in iPS cells.
Collapse
Affiliation(s)
- Song Park
- School of Life Science, BK21 plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea.,Core Protein Resources Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Yonghun Sung
- School of Life Science, BK21 plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Jain Jeong
- School of Life Science, BK21 plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Minjee Choi
- School of Life Science, BK21 plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Jinhee Lee
- School of Life Science, BK21 plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Wookbong Kwon
- School of Life Science, BK21 plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Soyoung Jang
- School of Life Science, BK21 plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Si Jun Park
- School of Life Science, BK21 plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Jae Young Kim
- Department of Biochemistry, School of Dentistry, IHBR, Kyungpook National University, Daegu, South Korea
| | - Sung Hyun Kim
- China-US (Henan) Hormel Cancer Institute, No.127 Dongming Road, Zhengzhou, Henan 450008, China
| | - Duhak Yoon
- The School of Animal BT Science, Kyungpook National University, Sangju-si, Gyeongsangbuk-do, South Korea
| | - Zae Young Ryoo
- School of Life Science, BK21 plus KNU Creative Bio Research Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Myoung Ok Kim
- The School of Animal BT Science, Kyungpook National University, Sangju-si, Gyeongsangbuk-do, South Korea
| |
Collapse
|
30
|
Qi Y, Cao KX, Xing FC, Zhang CY, Huang Q, Wu K, Wen FB, Zhao S, Li X. High expression of MAGE-A9 is associated with unfavorable survival in esophageal squamous cell carcinoma. Oncol Lett 2017; 14:3415-3420. [PMID: 28927095 PMCID: PMC5588010 DOI: 10.3892/ol.2017.6614] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 04/24/2017] [Indexed: 11/07/2022] Open
Abstract
Melanoma-associated antigens (MAGEs) are a group of well-characterized members of the cancer/testis antigen family, which are expressed in a variety of malignant tumors. MAGE-A9, a subfamily of MAGE-As, has been studied in a number of types of cancer and have been associated with unfavorable survival outcome. However, the expression of MAGE-A9 in human esophageal squamous cell carcinoma (ESCC) and association of MAGE-A9 with the clinicopathological characteristics of ESCC, particularly prognostic characteristics, remains unknown. The present study aimed at determining the expression level of MAGE-A9 and at evaluating its clinical significance in human ESCC. Quantitative polymerase chain reaction (qPCR) and immunohistochemistry (IHC) analyses were performed to characterize the expression of MAGE-A9 in ESCC tissues. Kaplan-Meier estimator survival and Coxs regression analyses were used to evaluate the prognosis of 103 patients with ESCC. The results of qPCR and IHC analysis revealed that the expression of MAGE-A9 was significantly increased in ESCC tissues, compared with that in healthy tissues. Furthermore, the expression level of MAGE-A9 protein in ESCC was significantly associated with the pathological grade (P=0.008), tumor size (P=0.027) and lymph node metastasis (P=0.009). Multivariate analysis using Coxs regression model identified that the expression level of MAGE-A9 and lymph node metastasis were independent prognostic factors for the overall survival rate of patients with ESCC (P=0.006 and P=0.001, respectively). The results of the present study are, to the best of our knowledge, the first to indicate that MAGE-A9 expression is a valuable prognostic biomarker for ESCC and that it may serve as a targeted therapy in the treatment of ESCC. Increased expression of MAGE-A9 indicated an unfavorable survival outcome in patients with ESCC.
Collapse
Affiliation(s)
- Yu Qi
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Ke Xin Cao
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Fu Chen Xing
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Chun Yang Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Qi Huang
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Kai Wu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Feng Biao Wen
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Song Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Xin Li
- Department of Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
31
|
Laiseca JE, Ladelfa MF, Cotignola J, Peche LY, Pascucci FA, Castaño BA, Galigniana MD, Schneider C, Monte M. Functional interaction between co-expressed MAGE-A proteins. PLoS One 2017; 12:e0178370. [PMID: 28542476 PMCID: PMC5443569 DOI: 10.1371/journal.pone.0178370] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 05/11/2017] [Indexed: 12/19/2022] Open
Abstract
MAGE-A (Melanoma Antigen Genes-A) are tumor-associated proteins with expression in a broad spectrum of human tumors and normal germ cells. MAGE-A gene expression and function are being increasingly investigated to better understand the mechanisms by which MAGE proteins collaborate in tumorigenesis and whether their detection could be useful for disease prognosis purposes. Alterations in epigenetic mechanisms involved in MAGE gene silencing cause their frequent co-expression in tumor cells. Here, we have analyzed the effect of MAGE-A gene co-expression and our results suggest that MageA6 can potentiate the androgen receptor (AR) co-activation function of MageA11. Database search confirmed that MageA11 and MageA6 are co-expressed in human prostate cancer samples. We demonstrate that MageA6 and MageA11 form a protein complex resulting in the stabilization of MageA11 and consequently the enhancement of AR activity. The mechanism involves association of the Mage A6-MHD domain to MageA11, prevention of MageA11 ubiquitinylation on lysines 240 and 245 and decreased proteasome-dependent degradation. We experimentally demonstrate here for the first time that two MAGE-A proteins can act together in a non-redundant way to potentiate a specific oncogenic function. Overall, our results highlight the complexity of the MAGE gene networking in regulating cancer cell behavior.
Collapse
Affiliation(s)
- Julieta E. Laiseca
- Lab. Oncología Molecular, Departamento de Química Biológica and IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María F. Ladelfa
- Lab. Oncología Molecular, Departamento de Química Biológica and IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Javier Cotignola
- Lab. Inflamación y Cáncer, Departamento de Química Biológica and IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Leticia Y. Peche
- Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, Area Science Park, Trieste , Italy
| | - Franco A. Pascucci
- Lab. Oncología Molecular, Departamento de Química Biológica and IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Bryan A. Castaño
- Lab. Oncología Molecular, Departamento de Química Biológica and IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mario D. Galigniana
- Lab. Biología Molecular y Celular, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Laboratorio Receptores Nucleares, IBYME-CONICET, Buenos Aires, Argentina
| | - Claudio Schneider
- Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, Area Science Park, Trieste , Italy
- Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, p.le Kolbe 4, Udine, Italy
| | - Martin Monte
- Lab. Oncología Molecular, Departamento de Química Biológica and IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
32
|
Mendonça BDS, Agostini M, Aquino IG, Dias WB, Bastos DC, Rumjanek FD. Suppression of MAGE-A10 alters the metastatic phenotype of tongue squamous cell carcinoma cells. Biochem Biophys Rep 2017; 10:267-275. [PMID: 28955754 PMCID: PMC5614724 DOI: 10.1016/j.bbrep.2017.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/13/2017] [Accepted: 04/18/2017] [Indexed: 12/21/2022] Open
Abstract
MAGE-A10 is a member of the MAGE protein family (melanoma associated antigen) which is overexpressed in cancer cells. Although MAGE-A10 has been characterized for some time and is generally associated to metastasis its function remains unknown. Here we describe experiments using as models oral squamous cell carcinoma (OSCC) cell lines displaying increasing metastatic potential (LN1 and LN2). These cell lines were transduced with lentivirus particles coding for short hairpin against MAGE-A10 mRNA. Repression of MAGE-A10 expression in LN2 cells altered their morphology and impaired growth of LN1 and LN2 cell lines. Furthermore, repression of MAGE-A10 expression increased cell-cell and cell matrix adhesion. Furthermore shMAGEA10 cells were shown to assemble aberrantly on a 3D culture system (microspheroids) when compared to cells transduced with the control scrambled construct. Cell migration was inhibited in knocked down cells as revealed by two different migration assays, wound healing and a phagokinetic track motility assay. In vitro invasion assay using a leiomyoma tissue derived matrix (myogel) showed that shMAGEA10 LN1 and shMAGEA10 LN2 cells displayed a significantly diminished ability to penetrate the matrices. Concomitantly, the expression of E-cadherin, N-cadherin and vimentin genes was analyzed. shMAGEA10 activated the expression of E-cadherin and repression N-cadherin and vimentin transcription. Taken together the results indicate that MAGE-A10 exerts its effects at the level of the epithelial-mesenchymal transition (EMT) presumably by regulating the expression of adhesion molecules.
Collapse
Affiliation(s)
- Bruna dos Santos Mendonça
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro Ilha do Fundão CEP 21941-902 Rio de Janeiro, Brazil
| | - Michelle Agostini
- Departamento de Patologia e Diagnóstico Oral - Faculdade de Odontologia, Universidade Federal do Rio de Janeiro, Brazil
| | - Iara Gonçalves Aquino
- Departamento de Patologia e Diagnóstico Oral - Faculdade de Odontologia, Universidade Federal do Rio de Janeiro, Brazil
| | - Wagner Barbosa Dias
- Laboratório de Glicobiologia Estrutural e Funcional Instituto de Biofísica-Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Débora Campanella Bastos
- Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas, Piracicaba, SP, Brazil
| | - Franklin D. Rumjanek
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro Ilha do Fundão CEP 21941-902 Rio de Janeiro, Brazil
- Corresponding author.
| |
Collapse
|
33
|
Demirbakan B, Sezgintürk MK. A sensitive and disposable indium tin oxide based electrochemical immunosensor for label-free detection of MAGE-1. Talanta 2017; 169:163-169. [PMID: 28411807 DOI: 10.1016/j.talanta.2017.03.076] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/20/2017] [Accepted: 03/25/2017] [Indexed: 01/21/2023]
Abstract
MAGE-1 (MAGE, for melanoma antigen), was identified by virtue of its processing and cell surface expression as a tumor-specific peptide bound to major histocompatibility complexes which was reactive with autolytic T cells. 3-Glycidoxypropyltrimethoxysilane (3-GOPS) is frequently employed for the preparation of dense heterometal hybrid polymers which are used, e.g., for hard coatings of organic polymers and contact lens materials in the optical industry. In this study, we have improved a new immunological biosensor with indium tin oxide (ITO). Then, Anti-MAGE-1 antibody was covalently immobilized with 3-GOPS which formed a self-assembled monolayers (SAMs) on modified ITO electrodes. Analytical characteristics such as square wave voltammetry, linear determination range, repeatability, reproducibility and regeneration of biosensors are determined. All characterization steps are monitored by electrochemical impedance spectroscopy (EIS), cyclic voltammetry (CV). The developed biosensor has wide determination range (0.5fg-15fg/mL). To investigate long shelf life of the fabricated biosensor, the immunosensors were stored at 4°C for periods ten weeks. Futhermore, binding kinetics of MAGE1 to antiMAGE-1 is monitored by single frequency technique in real time. Additionally, Kramer's-Kronig transform was used to understand whether the impedance spectra of biosensor system are affected from the variation that occurred because of external factor. Morphological characteristics of constructed biosensor were observed by scanning electron microscopy. Real human serum samples were also analyzed by the proposed biosensor, successfully. A commercial ELISA kit was also used as a reference method to validate the results obtained by the biosensor. Finally, this biosensor was tried in real blood sample and that showed it could be utilized in clinical applications. This biosensor can be preferred due to it has a wide linear range and it can be prepared easily.
Collapse
Affiliation(s)
- Burçak Demirbakan
- Namık Kemal University, Faculty of Science, Chemistry Department, Biochemistry Division, Tekirdağ, Turkey.
| | - Mustafa Kemal Sezgintürk
- Çanakkale Eighteen March University, Faculty of Engineering, Bioengineering Department, Çanakkale, Turkey.
| |
Collapse
|
34
|
Mecklenburg I, Sienel W, Schmid S, Passlick B, Kufer P. A Threshold of Systemic MAGE-A Gene Expression Predicting Survival in Resected Non-Small Cell Lung Cancer. Clin Cancer Res 2016; 23:1213-1219. [PMID: 27542766 DOI: 10.1158/1078-0432.ccr-16-0557] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 07/20/2016] [Accepted: 07/27/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Quantitative measurement of minimal residual disease predicting recurrence in individual cancer patients is available only in very few indications, such as acute lymphoblastic leukemia, but is still missing in most solid tumors, including non-small cell lung cancer (NSCLC).Experimental Design: MAGE-A expression levels in blood and bone marrow determined as calibrator-normalized relative ratios by quantitative multimarker real-time RT-PCR for transcript amplification of MAGE-A1, -A2, -A3/6, -A4, -A10, and -A12 in 94 patients with completely resected NSCLC were correlated with survival in a clinical study.Results: Patients with MAGE-A expression levels ≥0.2 in at least one sample of bone marrow or blood at tumor surgery had a significantly reduced overall (P = 0.007), cancer-free (P = 0.002), and distant metastasis-free survival (P < 0.001) versus patients below 0.2 in all samples without significant difference in locoregional recurrence-free survival. The corresponding HRs (≥0.2 vs. <0.2) for death, cancer-related death, and development of distant metastasis were 2.56 [95% confidence interval (CI), 1.42-4.63], 3.32 (95% CI, 1.66-6.61), and 4.03 (95% CI, 1.77-9.18), respectively. Five-year Kaplan-Meier estimates of distant metastasis-free survival were 43% (MAGE-A ≥ 0.2) versus 87% (MAGE-A < 0.2).Conclusions: MAGE-A expression in blood or bone marrow at tumor surgery is an independent predictor of survival in resected NSCLC. The reliable prediction of distant metastasis in individual patients with a statistically proven impact on overall survival may help to refine patient selection for adjuvant therapy urgently needed, especially in the clinical management of elderly patients. Clin Cancer Res; 23(5); 1213-9. ©2016 AACR.
Collapse
Affiliation(s)
- Ingo Mecklenburg
- Department of Internal Medicine, Klinikum Landsberg, Landsberg am Lech, Germany.
| | - Wulf Sienel
- Department of Thoracic Surgery, University Hospital Freiburg, Freiburg, Germany
| | - Severin Schmid
- Department of Thoracic Surgery, University Hospital Freiburg, Freiburg, Germany
| | - Bernward Passlick
- Department of Thoracic Surgery, University Hospital Freiburg, Freiburg, Germany
| | | |
Collapse
|
35
|
Biochemical and proteomic characterization of retrovirus Gag based microparticles carrying melanoma antigens. Sci Rep 2016; 6:29425. [PMID: 27403717 PMCID: PMC4941533 DOI: 10.1038/srep29425] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 06/17/2016] [Indexed: 12/22/2022] Open
Abstract
Extracellular vesicles are membraneous particles released by a variety of cells into the extracellular microenvironment. Retroviruses utilize the cellular vesiculation pathway for virus budding/assembly and the retrovirus Gag protein induces the spontaneous formation of microvesicles or virus-like particles (VLPs) when expressed in the mammalian cells. In this study, five different melanoma antigens, MAGEA4, MAGEA10, MART1, TRP1 and MCAM, were incorporated into the VLPs and their localization within the particles was determined. Our data show that the MAGEA4 and MAGEA10 proteins as well as MCAM are expressed on the surface of VLPs. The compartmentalization of exogenously expressed cancer antigens within the VLPs did not depend on the localization of the protein within the cell. Comparison of the protein content of VLPs by LC-MS/MS-based label-free quantitative proteomics showed that VLPs carrying different cancer antigens are very similar to each other, but differ to some extent from VLPs without recombinant antigen. We suggest that retrovirus Gag based virus-like particles carrying recombinant antigens have a potential to be used in cancer immunotherapy.
Collapse
|
36
|
Gao Y, Mutter-Rottmayer E, Greenwalt AM, Goldfarb D, Yan F, Yang Y, Martinez-Chacin RC, Pearce KH, Tateishi S, Major MB, Vaziri C. A neomorphic cancer cell-specific role of MAGE-A4 in trans-lesion synthesis. Nat Commun 2016; 7:12105. [PMID: 27377895 PMCID: PMC4935975 DOI: 10.1038/ncomms12105] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 05/31/2016] [Indexed: 12/11/2022] Open
Abstract
Trans-lesion synthesis (TLS) is an important DNA-damage tolerance mechanism that permits ongoing DNA synthesis in cells harbouring damaged genomes. The E3 ubiquitin ligase RAD18 activates TLS by promoting recruitment of Y-family DNA polymerases to sites of DNA-damage-induced replication fork stalling. Here we identify the cancer/testes antigen melanoma antigen-A4 (MAGE-A4) as a tumour cell-specific RAD18-binding partner and an activator of TLS. MAGE-A4 depletion from MAGE-A4-expressing cancer cells destabilizes RAD18. Conversely, ectopic expression of MAGE-A4 (in cell lines lacking endogenous MAGE-A4) promotes RAD18 stability. DNA-damage-induced mono-ubiquitination of the RAD18 substrate PCNA is attenuated by MAGE-A4 silencing. MAGE-A4-depleted cells fail to resume DNA synthesis normally following ultraviolet irradiation and accumulate γH2AX, thereby recapitulating major hallmarks of TLS deficiency. Taken together, these results demonstrate a mechanism by which reprogramming of ubiquitin signalling in cancer cells can influence DNA damage tolerance and probably contribute to an altered genomic landscape.
Collapse
Affiliation(s)
- Yanzhe Gao
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 101 Manning Drive, 614 Brinkhous-Bullitt Building, Chapel Hill, North Carolina 27599, USA
| | - Elizabeth Mutter-Rottmayer
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 101 Manning Drive, 614 Brinkhous-Bullitt Building, Chapel Hill, North Carolina 27599, USA
- Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Alicia M. Greenwalt
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 101 Manning Drive, 614 Brinkhous-Bullitt Building, Chapel Hill, North Carolina 27599, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Dennis Goldfarb
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Feng Yan
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Yang Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 101 Manning Drive, 614 Brinkhous-Bullitt Building, Chapel Hill, North Carolina 27599, USA
| | - Raquel C. Martinez-Chacin
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 101 Manning Drive, 614 Brinkhous-Bullitt Building, Chapel Hill, North Carolina 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Kenneth H. Pearce
- Center For Integrative Chemical Biology and Drug Discovery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Satoshi Tateishi
- Division of Cell Maintenance, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Honjo 2-2-1, Kumamoto 860-0811, Japan
| | - Michael B. Major
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Cyrus Vaziri
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 101 Manning Drive, 614 Brinkhous-Bullitt Building, Chapel Hill, North Carolina 27599, USA
- Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
37
|
MAGE-A1-6 expression in patients with head and neck squamous cell carcinoma: impact on clinical patterns and oncologic outcomes. Int J Clin Oncol 2016; 21:875-882. [PMID: 27230992 DOI: 10.1007/s10147-016-0989-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 05/03/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Various subtypes of melanoma-associated antigens (MAGEs) are expressed in the tumor tissues of patients with head and neck squamous cell carcinoma (HNSCC). However, little data are currently available on how the gene expression of MAGEs impacts clinical patterns and oncologic outcomes. We have therefore evaluated the expression of MAGE-A1-6 (A1-6) subtypes in tumor tissues of patients with HNSCC and the clinical impact of this expression. METHODS This was a retrospective review of 53 patients with histologically proven HNSCC of the oral cavity, oropharynx, larynx, or hypopharynx who underwent both treatment and analysis by reverse transcription (RT)-PCR assay with a common primer to identify the expression of MAGE-A1-6 subtypes in the tumor tissue. The clinicopathologic factors and oncologic outcomes of these patients and the correlations of both to MAGE-A1-6 gene expression were analyzed. RESULTS MAGE-A1-6 subtypes were expressed in the tumor tissues of 37 patients (69.8 %). Patient age of ≥65 years [p = 0.031, hazard ratio (HR) 4.866] and advanced American Joint Committee on Cancer stage (p = 0.035, HR 4.291) were independent risk factors for expression of MAGE-A1-6 subtypes. Patients with MAGE-A1-6 expression had lower disease-free survival (p = 0.029), disease-specific survival (p = 0.070), and overall survival (p = 0.017) rates. Overall survival rate was independently associated to chemotherapy (p = 0.011, HR 2.859), while no surgery (p = 0.050, HR 2.400) and MAGE-A1-6 expression (p = 0.050, HR 2.527) showed borderline significance. CONCLUSION In our patient group the expression of MAGE-A1-6 subtypes in tumor tissues of patients with HNSCC was correlated with advanced clinical stage of cancer and poor oncologic outcomes. We suggest that gene expression of MAGE-A1-6 subtypes may be considered to be a predictive factor to determine patient treatment or follow-up strategy.
Collapse
|
38
|
Pirlot C, Thiry M, Trussart C, Di Valentin E, Piette J, Habraken Y. Melanoma antigen-D2: A nucleolar protein undergoing delocalization during cell cycle and after cellular stress. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:581-95. [DOI: 10.1016/j.bbamcr.2015.12.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/11/2015] [Accepted: 12/15/2015] [Indexed: 12/25/2022]
|
39
|
Zang Y, Wei Y, Shi Y, Chen Q, Xing D. Chemo/Photoacoustic Dual Therapy with mRNA-Triggered DOX Release and Photoinduced Shockwave Based on a DNA-Gold Nanoplatform. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:756-769. [PMID: 26683002 DOI: 10.1002/smll.201502857] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/08/2015] [Indexed: 06/05/2023]
Abstract
A multifunctional nanoparticle based on gold nanorod (GNR), utilizing mRNA triggered chemo-drug release and near-infrared photoacoustic effect, is developed for a combined chemo-photoacoustic therapy. The constructed nanoparticle (GNR-DNA/FA:DOX) comprises three functional components: (i) GNR as the drug delivery platform and photoacoustic effect enhancer; (ii) toehold-possessed DNA dressed on the GNR to load doxorubicin (DOX) to implement a tumor cell specific chemotherapy; and (iii) folate acid (FA) modified on GNR to guide the nanoparticle to target tumor cells. The results show that, upon an effective and specific delivery of the nanoparticles to the tumor cells with overexpressed folate receptors, the cytotoxic DOX loaded on the GNR-DNA nanoplatform can be released through DNA displacement reaction in melanoma-associated antigen gene mRNA expressed cells. With 808 nm pulse laser irradiation, the photoacoustic effect of the GNR leads to a direct physical damage to the cells. The combined treatment of the two modalities can effectively destroy tumor cells and eradicate the tumors with two distinctively different and supplementing mechanisms. With the nanoparticle, photoacoustic imaging is successfully performed in situ to monitor the drug distribution and tumor morphology for therapeutical guidance. With further in-depth investigation, the proposed nanoparticle may provide an effective and safe alternative cancer treatment modality.
Collapse
Affiliation(s)
- Yundong Zang
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics South China Normal University, Guangzhou, 510631, P. R. China
| | - Yanchun Wei
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics South China Normal University, Guangzhou, 510631, P. R. China
| | - Yujiao Shi
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics South China Normal University, Guangzhou, 510631, P. R. China
| | - Qun Chen
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics South China Normal University, Guangzhou, 510631, P. R. China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics South China Normal University, Guangzhou, 510631, P. R. China
- Joint Laboratory of Laser Oncology with Cancer Center of Sun Yat-sen University, South China Normal University, Guangzhou, 510631, China
| |
Collapse
|
40
|
Liu S, Sang M, Xu Y, Gu L, Liu F, Shan B. Expression of MAGE-A1, -A9, -A11 in laryngeal squamous cell carcinoma and their prognostic significance: a retrospective clinical study. Acta Otolaryngol 2016; 136:506-13. [PMID: 26766421 DOI: 10.3109/00016489.2015.1126856] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONCLUSION The melanoma-associated antigens A1, -A9, -A11 (MAGE-A1, -A9, -A11) are relatively tumor-specific in laryngeal squamous cell carcinoma (LSCC), and could be ideal antigens for LSCC immunotherapy. In addition, MAGE-A9 probably is a poor prognostic marker for LSCC patients. OBJECTIVE The MAGE-A family belongs to Cancer/testis antigens (CTA). However, the expression pattern of MAGE-A1, MAGE-A9, and MAGE-A11 in LSCC is still unclear. This study aims to evaluate the expression and possible prognostic role of MAGE-A1, MAGE-A9, and MAGE-A11 in LSCC patients. METHODS The expression of MAGE-A1, MAGE-A9, and MAGE-A11 in LSCC specimens was investigated by immunohistochemistry, and the association of their expression and the clinical parameters and the survival of LSCC patients were analyzed by chi-square test, Kaplan-Meier survival and Cox regression analysis. RESULTS The expression rates of MAGE-A1, MAGE-A9, and MAGE-A11 in LSCC were 54.7%, 46.2%, and 51.9%, respectively. The expression of MAGE-A1, MAGE-A9, and MAGE-A11 in LSCC was correlated with clinical stage, lymph node metastasis, and tumor size. The overall survival of LSCC patients with positive MAGE-A1, MAGE-A9, or MAGE-A11 expression was lower than the patients without MAGE-A1, MAGE-A9, or MAGE-A11 expression. Cox's multivariable analysis showed that MAGE-A9 expression was an independently poor prognostic factor for LSCC patients.
Collapse
Affiliation(s)
- Shenghui Liu
- a Research Center, the Fourth Hospital of Hebei Medical University , Shijiazhuang , Hebei , PR China
- b Department of Otolaryngology , the Fourth Hospital of Hebei Medical University , Shijiazhuang , Hebei , PR China
| | - Meixiang Sang
- a Research Center, the Fourth Hospital of Hebei Medical University , Shijiazhuang , Hebei , PR China
| | - Yuru Xu
- b Department of Otolaryngology , the Fourth Hospital of Hebei Medical University , Shijiazhuang , Hebei , PR China
| | - Lina Gu
- a Research Center, the Fourth Hospital of Hebei Medical University , Shijiazhuang , Hebei , PR China
| | - Fei Liu
- a Research Center, the Fourth Hospital of Hebei Medical University , Shijiazhuang , Hebei , PR China
| | - Baoen Shan
- a Research Center, the Fourth Hospital of Hebei Medical University , Shijiazhuang , Hebei , PR China
| |
Collapse
|
41
|
Abstract
The past several years can be considered a renaissance era in the treatment of metastatic melanoma. Following a 30-year stretch in which oncologists barely put a dent in a very grim overall survival (OS) rate for these patients, things have rapidly changed course with the recent approval of three new melanoma drugs by the FDA. Both oncogene-targeted therapy and immune checkpoint blockade approaches have shown remarkable efficacy in a subset of melanoma patients and have clearly been game-changers in terms of clinical impact. However, most patients still succumb to their disease, and thus, there remains an urgent need to improve upon current therapies. Fortunately, innovations in molecular medicine have led to many silent gains that have greatly increased our understanding of the nature of cancer biology as well as the complex interactions between tumors and the immune system. They have also allowed for the first time a detailed understanding of an individual patient's cancer at the genomic and proteomic level. This information is now starting to be employed at all stages of cancer treatment, including diagnosis, choice of drug therapy, treatment monitoring, and analysis of resistance mechanisms upon recurrence. This new era of personalized medicine will foreseeably lead to paradigm shifts in immunotherapeutic treatment approaches such as individualized cancer vaccines and adoptive transfer of genetically modified T cells. Advances in xenograft technology will also allow for the testing of drug combinations using in vivo models, a truly necessary development as the number of new drugs needing to be tested is predicted to skyrocket in the coming years. This chapter will provide an overview of recent technological developments in cancer research, and how they are expected to impact future diagnosis, monitoring, and development of novel treatments for metastatic melanoma.
Collapse
Affiliation(s)
| | | | | | - Patrick Hwu
- University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Gregory Lizée
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
42
|
Kaushik A, Bhatia Y, Ali S, Gupta D. Gene Network Rewiring to Study Melanoma Stage Progression and Elements Essential for Driving Melanoma. PLoS One 2015; 10:e0142443. [PMID: 26558755 PMCID: PMC4641706 DOI: 10.1371/journal.pone.0142443] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/21/2015] [Indexed: 01/19/2023] Open
Abstract
Metastatic melanoma patients have a poor prognosis, mainly attributable to the underlying heterogeneity in melanoma driver genes and altered gene expression profiles. These characteristics of melanoma also make the development of drugs and identification of novel drug targets for metastatic melanoma a daunting task. Systems biology offers an alternative approach to re-explore the genes or gene sets that display dysregulated behaviour without being differentially expressed. In this study, we have performed systems biology studies to enhance our knowledge about the conserved property of disease genes or gene sets among mutually exclusive datasets representing melanoma progression. We meta-analysed 642 microarray samples to generate melanoma reconstructed networks representing four different stages of melanoma progression to extract genes with altered molecular circuitry wiring as compared to a normal cellular state. Intriguingly, a majority of the melanoma network-rewired genes are not differentially expressed and the disease genes involved in melanoma progression consistently modulate its activity by rewiring network connections. We found that the shortlisted disease genes in the study show strong and abnormal network connectivity, which enhances with the disease progression. Moreover, the deviated network properties of the disease gene sets allow ranking/prioritization of different enriched, dysregulated and conserved pathway terms in metastatic melanoma, in agreement with previous findings. Our analysis also reveals presence of distinct network hubs in different stages of metastasizing tumor for the same set of pathways in the statistically conserved gene sets. The study results are also presented as a freely available database at http://bioinfo.icgeb.res.in/m3db/. The web-based database resource consists of results from the analysis presented here, integrated with cytoscape web and user-friendly tools for visualization, retrieval and further analysis.
Collapse
Affiliation(s)
- Abhinav Kaushik
- Bioinformatics Laboratory, Structural and Computational Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India
| | - Yashuma Bhatia
- Bioinformatics Laboratory, Structural and Computational Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India
| | - Shakir Ali
- Department of Biochemistry, Faculty of Science, Jamia Hamdard, New Delhi, 110062, India
| | - Dinesh Gupta
- Bioinformatics Laboratory, Structural and Computational Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India
- * E-mail:
| |
Collapse
|
43
|
Wang CY, Lin BL, Chen CH. An aptamer targeting shared tumor-specific peptide antigen of MAGE-A3 in multiple cancers. Int J Cancer 2015; 138:918-26. [PMID: 26314689 DOI: 10.1002/ijc.29826] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 08/14/2015] [Indexed: 01/15/2023]
Abstract
A DNA aptamer was identified against the shared tumor-specific MAGE-A3111-125 peptide antigen. The dissociation constant between the aptamer and the peptide was measured at 57 nM. Binding of the aptamer to seven types of cancer cells, melanoma, breast, colorectal, liver, lung, pancreas and oral cancer, was confirmed with flow cytometry and fluorescence imaging. Cy3-conjugated aptamers signals were specifically localized to the surface of those cancer cells. The results indicate that the DNA aptamer against the shared tumor-specific MAGE-A3 peptide can be used in cancer cell targeting and has the potential for developing into new modalities for the diagnosis of multiple cancers.
Collapse
Affiliation(s)
- Chin-Yu Wang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Bai-Ling Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | |
Collapse
|
44
|
Li B, Zhu X, Sun L, Yuan L, Zhang J, Li H, Ye Z. Induction of a specific CD8+ T-cell response to cancer/testis antigens by demethylating pre-treatment against osteosarcoma. Oncotarget 2015; 5:10791-802. [PMID: 25301731 PMCID: PMC4279410 DOI: 10.18632/oncotarget.2505] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/16/2014] [Indexed: 01/18/2023] Open
Abstract
Conventional non-surgical therapeutic regimens against osteosarcoma are subject to chemoresistance and tumor relapse, and immunotherapy may be promising for this tumor. However, it's hard to find satisfactory epitopes for immunotherapy against osteosarcoma. Cancer/testis antigens (CTAs), such as MAGE-A family and NY-ESO-1, the potential antigens that almost exclusively express in tumor cells and immune-privileged sites, have been found expressed in osteosarcoma also. Nevertheless, the expression of CTAs is downregulated in many tumors, constraining the application of immunotherapy. In this article, we demonstrate that the expression of MAGE-A family and NY-ESO-1 in osteosarcoma cells can be upregulated following treatment with demethylating agent 5-aza-2'-deoxycytidine and consequently induces a CTA specific CD8+ T-cell response against osteosarcoma in vitro and in vivo. The in vivo imaging was realized by using luciferase-transfected HOS cells and DiR labeled T-cells in severely combined immunodeficiency mouse models. Cytotoxic T cells specifically recognizing MAGE-A family and NY-ESO-1 clustered at the tumor site in mice pre-treated with DAC and resulted in tumor growth suppression, while it was not observed in mice without DAC pre-treatment. This study is important for more targeted therapeutic approaches and suggests that adoptive immunotherapy, combined with demethylating treatment, has the potential for non-surgical therapeutic strategy against osteosarcoma.
Collapse
Affiliation(s)
- Binghao Li
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310008, China
| | - Xiaobing Zhu
- Department of Orthopaedics, Taizhou Cancer Hospital, Taizhou, 317502, China
| | - Lingling Sun
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310008, China
| | - Li Yuan
- School of Public Health, Fudan University, Shanghai, 200032, China
| | - Jian Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310008, China
| | - Hengyuan Li
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310008, China
| | - Zhaoming Ye
- Centre for Orthopaedic Research, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310008, China
| |
Collapse
|
45
|
Hou S, Sang M, Zhao L, Hou R, Shan B. The expression of MAGE-C1 and MAGE-C2 in breast cancer and their clinical significance. Am J Surg 2015; 211:142-51. [PMID: 26321295 DOI: 10.1016/j.amjsurg.2015.05.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 04/30/2015] [Accepted: 05/26/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND Our study aims to analyze the expression pattern, mechanism, and prognostic significance of melanoma-associated antigen MAGE-C1 and MAGE-C2 in breast cancer. METHODS Reverse transcription polymerase chain reaction (RT-PCR) and immunohistochemistry were used to investigate the expressions of MAGE-C1 and MAGE-C2 in breast benign disease specimens, tumor-free breast specimens, and breast cancer specimens; their correlation with clinicopathologic parameters and recurrence-free survival was elucidated. We examined the influence of DNA methylase inhibitor 5-aza-2'-deoxycytidine (5-aza-CdR) together with histone deacetylase inhibitor trichostatin A on the expression of MAGE-C1 and MAGE-C2 in breast cancer cell lines. RESULT Proteins for MAGE-C1 and MAGE-C2 expressions were 38.3% and 58.3% in breast cancer specimens, messenger RNA for MAGE-C1 and MAGE-C2 expressions were 43.3% and 61.7%, respectively. MAGE-C1 and MAGE-C2 expressions were positively associated with high tumor grade and reduced recurrence-free survival; MAGE-C2 expression was also associated with tumor embolus and histologic type. 5-aza-CdR treatment alone could induce expression of MAGE-C2, whereas trichostatin A was able to synergistically enhance 5-aza-CdR-mediated MAGE-C2 transcription. CONCLUSIONS MAGE-C1 and MAGE-C2 maybe potential targets for tumor immunotherapy, and their expressions are associated with advanced breast cancer and poor outcome.
Collapse
Affiliation(s)
- Shuyun Hou
- Department of Immunology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China; Research Center, The Fourth Hospital of Hebei Medical University, Jiankang Road 12th, Shijiazhuang 050017, Hebei, People's Republic of China
| | - Meixiang Sang
- Research Center, The Fourth Hospital of Hebei Medical University, Jiankang Road 12th, Shijiazhuang 050017, Hebei, People's Republic of China
| | - Lianmei Zhao
- Research Center, The Fourth Hospital of Hebei Medical University, Jiankang Road 12th, Shijiazhuang 050017, Hebei, People's Republic of China
| | - Ran Hou
- Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Baoen Shan
- Research Center, The Fourth Hospital of Hebei Medical University, Jiankang Road 12th, Shijiazhuang 050017, Hebei, People's Republic of China.
| |
Collapse
|
46
|
Hao MZ, Zhou WY, Du XL, Chen KX, Wang GW, Yang Y, Yang JL. Novel anti-melanoma treatment: focus on immunotherapy. CHINESE JOURNAL OF CANCER 2015; 33:458-65. [PMID: 25189718 PMCID: PMC4190435 DOI: 10.5732/cjc.014.10118] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Melanoma is an intractable cancer that is aggressive, lethal, and metastatic. The prognosis of advanced melanoma is very poor because it is insensitive to chemotherapy and radiotherapy. The incidence of melanoma has been ascending stably for years worldwide, accompanied by increasing mortality. New approaches to managing this deadly disease are much anticipated to enhance the cure rate and to extend clinical benefits to patients with metastatic melanoma. Due to its high degree of immunogenicity, melanoma could be a good target for immunotherapy, which has been developed for decades and has achieved certain progress. This article provides an overview of immunotherapy for melanoma.
Collapse
Affiliation(s)
- Meng-Ze Hao
- Department of Bone and Soft Tissue Tumor, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Hospital & Institute, Tianjin 30060, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
47
|
Marcar L, Ihrig B, Hourihan J, Bray SE, Quinlan PR, Jordan LB, Thompson AM, Hupp TR, Meek DW. MAGE-A Cancer/Testis Antigens Inhibit MDM2 Ubiquitylation Function and Promote Increased Levels of MDM4. PLoS One 2015; 10:e0127713. [PMID: 26001071 PMCID: PMC4441487 DOI: 10.1371/journal.pone.0127713] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 04/17/2015] [Indexed: 02/04/2023] Open
Abstract
Melanoma antigen A (MAGE-A) proteins comprise a structurally and biochemically similar sub-family of Cancer/Testis antigens that are expressed in many cancer types and are thought to contribute actively to malignancy. MAGE-A proteins are established regulators of certain cancer-associated transcription factors, including p53, and are activators of several RING finger-dependent ubiquitin E3 ligases. Here, we show that MAGE-A2 associates with MDM2, a ubiquitin E3 ligase that mediates ubiquitylation of more than 20 substrates including mainly p53, MDM2 itself, and MDM4, a potent p53 inhibitor and MDM2 partner that is structurally related to MDM2. We find that MAGE-A2 interacts with MDM2 via the N-terminal p53-binding pocket and the RING finger domain of MDM2 that is required for homo/hetero-dimerization and for E2 ligase interaction. Consistent with these data, we show that MAGE-A2 is a potent inhibitor of the E3 ubiquitin ligase activity of MDM2, yet it does not have any significant effect on p53 turnover mediated by MDM2. Strikingly, however, increased MAGE-A2 expression leads to reduced ubiquitylation and increased levels of MDM4. Similarly, silencing of endogenous MAGE-A expression diminishes MDM4 levels in a manner that can be rescued by the proteasomal inhibitor, bortezomid, and permits increased MDM2/MDM4 association. These data suggest that MAGE-A proteins can: (i) uncouple the ubiquitin ligase and degradation functions of MDM2; (ii) act as potent inhibitors of E3 ligase function; and (iii) regulate the turnover of MDM4. We also find an association between the presence of MAGE-A and increased MDM4 levels in primary breast cancer, suggesting that MAGE-A-dependent control of MDM4 levels has relevance to cancer clinically.
Collapse
Affiliation(s)
- Lynnette Marcar
- Division of Cancer Research, University of Dundee, Clinical Research Centre and Jacqui Wood Cancer Centre, Ninewells Hospital, James Arrott Drive, Dundee, United Kingdom
| | - Bianca Ihrig
- Division of Cancer Research, University of Dundee, Clinical Research Centre and Jacqui Wood Cancer Centre, Ninewells Hospital, James Arrott Drive, Dundee, United Kingdom
| | - John Hourihan
- Division of Cancer Research, University of Dundee, Clinical Research Centre and Jacqui Wood Cancer Centre, Ninewells Hospital, James Arrott Drive, Dundee, United Kingdom
| | - Susan E. Bray
- Division of Cancer Research, University of Dundee, Clinical Research Centre and Jacqui Wood Cancer Centre, Ninewells Hospital, James Arrott Drive, Dundee, United Kingdom
| | - Philip R. Quinlan
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, United Kingdom
- School of Computer Science, University of Nottingham, Jubilee Campus, Nottingham, United Kingdom
- Advanced Data Analysis Centre, University of Nottingham, Nottingham, United Kingdom
| | - Lee B. Jordan
- Division of Cancer Research, University of Dundee, Clinical Research Centre and Jacqui Wood Cancer Centre, Ninewells Hospital, James Arrott Drive, Dundee, United Kingdom
| | - Alastair M. Thompson
- M. D. Anderson Cancer Center, University of Texas, 1400 Pressler Drive, Unit 1484, Houston, United States of America
| | - Ted R. Hupp
- p53 Signal Transduction Laboratory, Edinburgh Cancer Research UK Centre, The University of Edinburgh, Crewe Road South, Edinburgh, United Kingdom
| | - David W. Meek
- Division of Cancer Research, University of Dundee, Clinical Research Centre and Jacqui Wood Cancer Centre, Ninewells Hospital, James Arrott Drive, Dundee, United Kingdom
- * E-mail:
| |
Collapse
|
48
|
Lu Z, Jiao D, Qiao J, Yang S, Yan M, Cui S, Liu Z. Restin suppressed epithelial-mesenchymal transition and tumor metastasis in breast cancer cells through upregulating mir-200a/b expression via association with p73. Mol Cancer 2015; 14:102. [PMID: 25972084 PMCID: PMC4429374 DOI: 10.1186/s12943-015-0370-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 04/21/2015] [Indexed: 12/31/2022] Open
Abstract
Background Restin belongs to MAGE superfamily and is known as MAGE H1. Restin was firstly cloned from HL-60 cells treated with all-trans retinoic acid (ATRA). Previous studies showed a pro-apoptotic role of Restin in several cell lines. However, little information is available on its expression patterns and functions in vivo. Our study was performed to detect if Restin plays a role in breast cancer cells in vitro and in vivo. Methods and results Real-time PCR and western blot were conducted to detect Restin expression in multiple breast cancer cell lines and Restin level was negatively related with cell motility. Restin overexpression and knockdown stable cell lines were established by transducing lentivirus into MCF-7 and MDA-MB-231 cells. Cell morphology, wound closure assay, transwell migration and invasion assays were performed to detect if Restin inhibited EMT. Our data showed that Restin overexpressed cells exhibited classical epithelial cell morphology, and Restin overexpression resulted in activation of epithelial markers and suppression of mesenchymal markers, and inhibition of cell migration and invasion. Tumor xenograft model was used to characterize the biological functions of Restin in vivo. We found that Restin overexpression led to reduced lung metastasis. Real-time PCR, western blot, luciferase assay and ChIP assay were performed to identify the potential targets of Restin and the underlying molecular mechanisms. Among several master regulators of EMT, only ZEB1/2 levels were dramatically inhibited by Restin. Unexpectedly, Restin indirectly regulated ZEB1/2 expression at post-transcriptional level. We further identified mir-200a/b, well-characterized mediators controlling ZEB1/2 expression, were transcriptionally activated by Restin and the regulation was dependent on the p53 binding site in mir-200b/a/429 promoter. Further mechanical studies demonstrated Restin interacted with p73, one of p53 family members, which contributed to Restin-mediated activation of mir-200a/b and suppression of ZEB1/2. Conclusions Taken together, our results suggest that Restin inhibits EMT and tumor metastasis by controlling the expression of the tumor metastasis suppressor mir-200a/b via association with p73. Our findings not only establish a mechanistic link between Restin, EMT and tumor metastasis, but also provide strong evidence supporting the notion that MAGE Group II proteins may exert a tumor suppressive effect in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0370-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhenduo Lu
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Dechuang Jiao
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Jianghua Qiao
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Sen Yang
- Department of Pathogen Biology, Basic Medical College of Zhengzhou University, #100 Science Road, Zhengzhou, 450001, People's Republic of China.
| | - Min Yan
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Shude Cui
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Zhenzhen Liu
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| |
Collapse
|
49
|
Xie C, Wang G. XAGE-1b cancer/testis antigen is a potential target for immunotherapy in prostate cancer. Urol Int 2015; 94:354-62. [PMID: 25572590 DOI: 10.1159/000363333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 04/29/2014] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Gene-modified cell vaccines are now considered to be the best way to achieve immunotherapy for a variety of cancers including prostate cancer (PCa). XAGE-1b is a member of the cancer/testis antigen family which has demonstrated strong immunogenicity. We investigated whether XAGE-1b is an ideal target for PCa immunotherapy. MATERIALS AND METHODS The recombinant eukaryotic expression vector pDisplay-XAGE-1b was constructed. Then the recombinant vector was transfected into Myc-CaP cells and its immunogenicity in vitro was studied. After transfection, the Myc-CaP-XAGE-1b cells were injected into FVB mice subcutaneously. Tumor growth was periodically observed and the anti-tumor effect and mechanism in vivo were further studied. RESULTS The recombinant vector was correctly constructed by DNA sequencing and restriction endonuclease digestion. Myc-CaP cells were successfully transfected with XAGE-1b gene by immunofluorescence staining and Western blot. The transfected cells exhibited increased IFN-γ secretion, decreased IL-6 secretion and enhanced killing activity. Tumor grew slower in XAGE-1b-modified FVB mice than in wild-type FVB mice. High dendritic cell expression and low myeloid-derived suppressor cell expression were observed in tumor tissues expressed with XAGE-1b. CONCLUSIONS XAGE-1b gene transfection could significantly enhance the immunogenicity of Myc-CaP cells. Therefore, XAGE-1b may be an attractive target for antigen-specific immunotherapy in PCa.
Collapse
Affiliation(s)
- Chong Xie
- Department of Andrology, International Peace Maternity and Child Health Hospital, Shanghai Jiaotong University, Shanghai, China
| | | |
Collapse
|
50
|
Zhang QM, Shen N, Xie S, Bi SQ, Luo B, Lin YD, Fu J, Zhou SF, Luo GR, Xie XX, Xiao SW. MAGED4 expression in glioma and upregulation in glioma cell lines with 5-aza-2'-deoxycytidine treatment. Asian Pac J Cancer Prev 2015; 15:3495-501. [PMID: 24870746 DOI: 10.7314/apjcp.2014.15.8.3495] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Melanoma-associated antigen (MAGE) family genes have been considered as potentially promising targets for anticancer immunotherapy. MAGED4 was originally identified as a glioma-specific antigen. Current knowledge about MAGED4 expression in glioma is only based on mRNA analysis and MAGED4 protein expression has not been elucidated. In the present study, we investigated this point and found that MAGED4 mRNA and protein were absent or very lowly expressed in various normal tissues and glioma cell line SHG44, but overexpressed in glioma cell lines A172,U251,U87-MG as well as glioma tissues, with significant heterogeneity. Furthermore, MAGED4 protein expression was positively correlated with the glioma type and grade. We also found that the expression of MAGED4 inversely correlated with the overall methylation status of the MAGED4 promoter CpG island. Furthermore, when SHG44 and A172 with higher methylation were treated with the DNA demethylating agent 5-aza-2'-deoxycytidine (5-AZA-CdR) reactivation of MAGED4 mRNA was mediated by significant demethylation in SHG44 instead of A172. However, 5-AZA-CdR treatment had no effect on MAGED4 protein in both SHG44 and A172 cells. In conclusion, MAGED4 is frequently and highly expressed in glioma and is partly regulated by DNA methylation. The results suggest that MAGED4 might be a promising target for glioma immunotherapy combined with 5-AZA-CdR to enhance its expression and eliminate intratumor heterogeneity.
Collapse
Affiliation(s)
- Qing-Mei Zhang
- Department of Histology and Embryology, School of Pre-clinical Medicine, Guangxi Medical University, Guangxi, China E-mail :
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|