1
|
Lin L, Zou X, Nong W, Ge Y, Li F, Luo B, Zhang Q, Xie X. The potential value of cancer-testis antigens in ovarian cancer: Prognostic markers and targets for immunotherapy. Immun Inflamm Dis 2024; 12:e1284. [PMID: 38896069 PMCID: PMC11186301 DOI: 10.1002/iid3.1284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/11/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Tumor immunotherapy has become an important adjuvant therapy after surgery, radiotherapy, and chemotherapy. In recent years, the role of tumor-associated antigen (TAA) in tumor immunotherapy has become increasingly prominent. Cancer-testis antigen (CTA) is a kind of TAA that is highly restricted in a variety of tumors and can induce an immune response. AIMS This review article aimed to evaluate the role of CTA on the progression of ovarian cancer, its diagnostic efficacy, and the potential for immunotherapy. METHODS We analyzed publications and outlined a comprehensive of overview the regulatory mechanism, immunogenicity, clinical expression significance, tumorigenesis, and application prospects of CTA in ovarian cancer, with a particular focus on recent progress in CTA-based immunotherapy. RESULTS The expression of CTA affects the occurrence, development, and prognosis of ovarian cancer and is closely related to tumor immunity. CONCLUSION CTA can be used as a biomarker for the diagnosis and prognosis evaluation of ovarian cancer and is an ideal target for antitumor immunotherapy. These findings provide novel insights on CTA in the improvement of diagnosis and treatment for ovarian cancer. The successes, current challenges and future prospects were also discussed to portray its significant potential.
Collapse
Affiliation(s)
- Lina Lin
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Xiaoqiong Zou
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Weixia Nong
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Yingying Ge
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Feng Li
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Bin Luo
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
- Education Department of Guangxi Zhuang Autonomous RegionKey Laboratory of Basic Research on Regional Diseases (Guangxi Medical University)NanningGuangxiPeople's Republic of China
| | - Qingmei Zhang
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
- Education Department of Guangxi Zhuang Autonomous RegionKey Laboratory of Basic Research on Regional Diseases (Guangxi Medical University)NanningGuangxiPeople's Republic of China
| | - Xiaoxun Xie
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
- Education Department of Guangxi Zhuang Autonomous RegionKey Laboratory of Basic Research on Regional Diseases (Guangxi Medical University)NanningGuangxiPeople's Republic of China
- Ministry of Education, Key Laboratory of Early Prevention and Treatment of Regional High Frequency Tumor (Guangxi Medical University)NanningGuangxiPeople's Republic of China
| |
Collapse
|
2
|
The Melanoma-Associated Antigen Family A (MAGE-A): A Promising Target for Cancer Immunotherapy? Cancers (Basel) 2023; 15:cancers15061779. [PMID: 36980665 PMCID: PMC10046478 DOI: 10.3390/cancers15061779] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023] Open
Abstract
Early efforts to identify tumor-associated antigens over the last decade have provided unique cancer epitopes for targeted cancer therapy. MAGE-A proteins are a subclass of cancer/testis (CT) antigens that are presented on the cell surface by MHC class I molecules as an immune-privileged site. This is due to their restricted expression to germline cells and a wide range of cancers, where they are associated with resistance to chemotherapy, metastasis, and cancer cells with an increasing potential for survival. This makes them an appealing candidate target for designing an effective and specific immunotherapy, thereby suggesting that targeting oncogenic MAGE-As with cancer vaccination, adoptive T-cell transfer, or a combination of therapies would be promising. In this review, we summarize and discuss previous and ongoing (pre-)clinical studies that target these antigens, while bearing in mind the benefits and drawbacks of various therapeutic strategies, in order to speculate on future directions for MAGE-A-specific immunotherapies.
Collapse
|
3
|
Saha C, Bojdo J, Dunne NJ, Duary RK, Buckley N, McCarthy HO. Nucleic acid vaccination strategies for ovarian cancer. Front Bioeng Biotechnol 2022; 10:953887. [PMID: 36420446 PMCID: PMC9677957 DOI: 10.3389/fbioe.2022.953887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 10/26/2022] [Indexed: 09/19/2023] Open
Abstract
High grade serous carcinoma (HGSC) is one of the most lethal ovarian cancers that is characterised by asymptomatic tumour growth, insufficient knowledge of malignant cell origin and sub-optimal detection. HGSC has been recently shown to originate in the fallopian tube and not in the ovaries. Conventional treatments such as chemotherapy and surgery depend upon the stage of the disease and have resulted in higher rates of relapse. Hence, there is a need for alternative treatments. Differential antigen expression levels have been utilised for early detection of the cancer and could be employed in vaccination strategies using nucleic acids. In this review the different vaccination strategies in Ovarian cancer are discussed and reviewed. Nucleic acid vaccination strategies have been proven to produce a higher CD8+ CTL response alongside CD4+ T-cell response when compared to other vaccination strategies and thus provide a good arena for antitumour immune therapy. DNA and mRNA need to be delivered into the intracellular matrix. To overcome ineffective naked delivery of the nucleic acid cargo, a suitable delivery system is required. This review also considers the suitability of cell penetrating peptides as a tool for nucleic acid vaccine delivery in ovarian cancer.
Collapse
Affiliation(s)
- Chayanika Saha
- School of Pharmacy, Queen’s University of Belfast, Belfast, United Kingdom
| | - James Bojdo
- School of Pharmacy, Queen’s University of Belfast, Belfast, United Kingdom
| | - Nicholas J. Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin, Ireland
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- Advanced Processing Technology Research Centre, Dublin City University, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Raj Kumar Duary
- Department of Food Engineering and Technology, Tezpur University, Tezpur, India
| | - Niamh Buckley
- School of Pharmacy, Queen’s University of Belfast, Belfast, United Kingdom
| | - Helen O. McCarthy
- School of Pharmacy, Queen’s University of Belfast, Belfast, United Kingdom
- School of Chemical Sciences, Dublin City University, Dublin, Ireland
| |
Collapse
|
4
|
Dai X, Li J, Chen Y, Ostrikov KK. When Onco-Immunotherapy Meets Cold Atmospheric Plasma: Implications on CAR-T Therapies. Front Oncol 2022; 12:837995. [PMID: 35280746 PMCID: PMC8905244 DOI: 10.3389/fonc.2022.837995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/31/2022] [Indexed: 12/03/2022] Open
Abstract
T cells engineered with chimeric antigen receptors (CAR) have demonstrated its widespread efficacy as a targeted immunotherapeutic modality. Yet, concerns on its specificity, efficacy and generalization prevented it from being established into a first-line approach against cancers. By reviewing challenges limiting its clinical application, ongoing efforts trying to resolve them, and opportunities that emerging oncotherapeutic modalities may bring to temper these challenges, we conclude that careful CAR design should be done to avoid the off-tumor effect, enhance the efficacy of solid tumor treatment, improve product comparability, and resolve problems such as differential efficacies of co-stimulatory molecules, cytokine storm, tumor lysis syndrome, myelosuppression and severe hepatotoxicity. As a promising solution, we propose potential synergies between CAR-T therapies and cold atmospheric plasma, an emerging onco-therapeutic strategy relying on reactive species, towards improved therapeutic efficacies and enhanced safety that deserve extensive investigations.
Collapse
Affiliation(s)
- Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.,CAPsoul Biotechnology Company, Ltd, Beijing, China
| | - Jitian Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Provincial Orthopedic Institute, Zhengzhou, China
| | - Yiming Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Kostya Ken Ostrikov
- School of Chemistry and Physics and Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
5
|
Poplawska M, Dutta D, Lee Y, Lim SH. Sperm protein 17 targeting for epithelial ovarian cancer treatment in the era of modern immunoengineering. Mol Ther Oncolytics 2021; 23:378-386. [PMID: 34853809 PMCID: PMC8604669 DOI: 10.1016/j.omto.2021.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
6
|
Oncogenic activity and cellular functionality of melanoma associated antigen A3. Biochem Pharmacol 2021; 192:114700. [PMID: 34303709 DOI: 10.1016/j.bcp.2021.114700] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022]
Abstract
Cancer testis antigen Melanoma associated antigen A3 (MAGE-A3) has been subject of research for many years. Being expressed in various tumor types and influencing proliferation, metastasis, and tumor pathogenicity, MAGE-A3 is an attractive target for cancer therapy, particularly because in healthy tissues, MAGE-A3 is only expressed in testes and placenta. MAGE-A3 acts as a cellular master regulator by stimulating E3 ubiquitin ligase tripartite motif-containing protein 28 (TRIM28), resulting in regulation of various cellular targets. These include tumor suppressor protein p53 and cellular energy sensor AMP-activated protein kinase (AMPK). The restricted expression of MAGE-A3 in tumor cells makes MAGE-A3 an attractive target for vaccine-based immune therapy. However, although phase I and phase II clinical trials involving MAGE-A3-specific immunotherapeutic interventions were promising, large phase III studies failed. This article gives an overview about the role of MAGE-A3 as a cellular master switch and discusses approaches to improve MAGE-A3-based immunotherapies.
Collapse
|
7
|
Batchu RB, Gruzdyn OV, Kolli BK, Dachepalli R, Umar PS, Rai SK, Singh N, Tavva PS, Weaver DW, Gruber SA. IL-10 Signaling in the Tumor Microenvironment of Ovarian Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1290:51-65. [PMID: 33559854 DOI: 10.1007/978-3-030-55617-4_3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Unlike other malignancies, ovarian cancer (OC) creates a complex tumor microenvironment with distinctive peritoneal ascites consisting of a mixture of several immunosuppressive cells which impair the ability of the patient's immune system to fight the disease. The poor survival rates observed in advanced stage OC patients and the lack of effective conventional therapeutic options have been attributed in large part to the immature dendritic cells (DCs), IL-10 secreting regulatory T cells, tumor-associated macrophages, myeloid-derived suppressor cells, and cancer stem cells that secrete inhibitory cytokines. This review highlights the critical role played by the intraperitoneal presence of IL-10 in the generation of an immunosuppressive tumor microenvironment. Further, the effect of antibody neutralization of IL-10 on the efficacy of DC and chimeric antigen receptor T-cell vaccines will be discussed. Moreover, we will review the influence of IL-10 in the promotion of cancer stemness in concert with the NF-κB signaling pathway with regard to OC progression. Finally, understanding the role of IL-10 and its crosstalk with various cells in the ascitic fluid may contribute to the development of novel immunotherapeutic approaches with the potential to kill drug-resistant OC cells while minimizing toxic side effects.
Collapse
Affiliation(s)
- Ramesh B Batchu
- Wayne State University School of Medicine, Detroit, MI, USA. .,John D. Dingell VA Medical Center, Detroit, MI, USA.
| | - Oksana V Gruzdyn
- Wayne State University School of Medicine, Detroit, MI, USA.,John D. Dingell VA Medical Center, Detroit, MI, USA
| | - Bala K Kolli
- Wayne State University School of Medicine, Detroit, MI, USA.,John D. Dingell VA Medical Center, Detroit, MI, USA.,Med Manor Organics Pvt. Ltd., Hyderabad, India
| | | | - Prem S Umar
- Med Manor Organics Pvt. Ltd., Hyderabad, India
| | | | | | | | | | - Scott A Gruber
- Wayne State University School of Medicine, Detroit, MI, USA.,John D. Dingell VA Medical Center, Detroit, MI, USA
| |
Collapse
|
8
|
Das B, Senapati S. Immunological and functional aspects of MAGEA3 cancer/testis antigen. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 125:121-147. [PMID: 33931137 DOI: 10.1016/bs.apcsb.2020.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Identification of ectopic gene activation in cancer cells serves as a basis for both gene signature-guided tumor targeting and unearthing of oncogenic mechanisms to expand the understanding of tumor biology/oncogenic process. Proteins expressed only in germ cells of testis and/or placenta (immunoprivileged organs) and in malignancies are called cancer testis antigens; they are antigenic because of the lack of antigen presentation by those specific cell types (germ cells), which limits the exposure of the proteins to the immune cells. Since the Cancer Testis Antigens (CTAs) are immunogenic and expressed in a wide variety of cancer types, CT antigens have become interesting target for immunotherapy against cancer. Among CT antigens MAGEA family is reported to have 12 members (MAGEA1 to MAGEA12). The current review highlights the studies on MAGEA3 which is a CT antigen and reported in almost all types of cancer. MAGEA3 is well tried for cancer immunotherapy. Recent advances on its functional and immunological aspect warranted much deliberation on effective therapeutic approach, thus making it a more interesting target for cancer therapy.
Collapse
Affiliation(s)
- Biswajit Das
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India; Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shantibhusan Senapati
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India.
| |
Collapse
|
9
|
Taherian-Esfahani Z, Dashti S. Cancer-testis antigens: An update on their roles in cancer immunotherapy. Hum Antibodies 2020; 27:171-183. [PMID: 30909205 DOI: 10.3233/hab-190366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Several recent studies have assessed suitability of tumor antigens for immunotherapy. Based on the restricted expression pattern in somatic tissues, cancer-testis antigens (CTAs) are possible candidates for cancer immunotherapy. These antigens are expressed in various tumors including gastrointestinal, breast, skin and hematologic malignancies. OBJECTIVES To find clinical trials utilizing CTAs in cancer patients. METHODS We searched PubMed, google scholar and specific websites that registers clinical trials. RESULTS A number of clinical trials have been designed to evaluate safety and efficacy of CTA-based treatments. The results of some of them have been promising. In the current literature search, we summarized the clinical trials of CTA-based therapies in cancer patients. CONCLUSIONS Based on the availability of different formulations of CTA-based vaccines, future researches should compare efficiency of these modalities.
Collapse
|
10
|
Zebularine Treatment Induces MAGE-A11 Expression and Improves CTL Cytotoxicity Using a Novel Identified HLA-A2-restricted MAGE-A11 Peptide. J Immunother 2017; 40:211-220. [DOI: 10.1097/cji.0000000000000170] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
11
|
Zajac P, Schultz-Thater E, Tornillo L, Sadowski C, Trella E, Mengus C, Iezzi G, Spagnoli GC. MAGE-A Antigens and Cancer Immunotherapy. Front Med (Lausanne) 2017; 4:18. [PMID: 28337438 PMCID: PMC5340762 DOI: 10.3389/fmed.2017.00018] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 02/09/2017] [Indexed: 12/30/2022] Open
Abstract
MAGE-A antigens are expressed in a variety of cancers of diverse histological origin and germinal cells. Due to their relatively high tumor specificity, they represent attractive targets for active specific and adoptive cancer immunotherapies. Here, we (i) review past and ongoing clinical studies targeting these antigens, (ii) analyze advantages and disadvantages of different therapeutic approaches, and (iii) discuss possible improvements in MAGE-A-specific immunotherapies.
Collapse
Affiliation(s)
- Paul Zajac
- Oncology Surgery, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| | - Elke Schultz-Thater
- Oncology Surgery, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| | - Luigi Tornillo
- Department of Pathology, University Hospital of Basel , Basel , Switzerland
| | - Charlotte Sadowski
- Oncology Surgery, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| | - Emanuele Trella
- Oncology Surgery, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| | - Chantal Mengus
- Oncology Surgery, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| | - Giandomenica Iezzi
- Cancer Immunotherapy, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| | - Giulio C Spagnoli
- Oncology Surgery, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| |
Collapse
|
12
|
Taherian-Esfahani Z, Abedin-Do A, Nikpayam E, Tasharofi B, Ghahghaei Nezamabadi A, Ghafouri-Fard S. Cancer-Testis Antigens: A Novel Group of Tumor Biomarkers in Ovarian Cancers. IRANIAN JOURNAL OF CANCER PREVENTION 2016. [DOI: 10.17795/ijcp-4993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
13
|
Immune effect and safety evaluation of vaccine prepared by dendritic cells modified by rAAV-carrying BCSG1 gene. Gene Ther 2016; 23:839-845. [PMID: 27556816 DOI: 10.1038/gt.2016.63] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/17/2016] [Accepted: 08/01/2016] [Indexed: 12/24/2022]
Abstract
The immune effect and safety evaluation of rAAV (recombinant adeno-associated virus)-containing Bcsg1 (breast cancer-specific gene 1) (rAAV/Bcsg1)-transfected DC (dendritic cell) (rAAV/Bcsg1-DC) vaccine in immunotherapy for (BCSG1) (+) BC was assessed. Immune effect of cytotoxic T lymphocytes (CTLs) on Bcsg1 (+) BC cells, and rAAV gene residuals in mature CTL cells and culture medium were determined. Nude mouse xenograft tumor model was established to assess the inhibition effects of DC-activated CTLs on tumor growth. DC cell surface markers were highly expressed in rAAV/Bcsg1 group and lysate-DC group, and rAAV/Bcsg1-DC-CTL showed stronger cytotoxic activity targeting Bcsg1 (+) BC cells. The rAAV/Bcsg1-DC vaccine-treated groups showed lower mean tumor weight, higher tumor inhibition rates and slower tumor growth. rAAV/Bcsg1-DC can induce highly efficient CTL-targeting Bcsg1 antigen without rAAV gene residuals.
Collapse
|
14
|
Chang KP, Kolli BK. New "light" for one-world approach toward safe and effective control of animal diseases and insect vectors from leishmaniac perspectives. Parasit Vectors 2016; 9:396. [PMID: 27412129 PMCID: PMC4942964 DOI: 10.1186/s13071-016-1674-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/29/2016] [Indexed: 01/03/2023] Open
Abstract
Light is known to excite photosensitizers (PS) to produce cytotoxic reactive oxygen species (ROS) in the presence of oxygen. This modality is attractive for designing control measures against animal diseases and pests. Many PS have a proven safety record. Also, the ROS cytotoxicity selects no resistant mutants, unlike other drugs and pesticides. Photodynamic therapy (PDT) refers to the use of PS as light activable tumoricides, microbicides and pesticides in medicine and agriculture. Here we describe “photodynamic vaccination” (PDV) that uses PDT-inactivation of parasites, i.e. Leishmania as whole-cell vaccines against leishmaniasis, and as a universal carrier to deliver transgenic add-on vaccines against other infectious and malignant diseases. The efficacy of Leishmania for vaccine delivery makes use of their inherent attributes to parasitize antigen (vaccine)-presenting cells. Inactivation of Leishmania by PDT provides safety for their use. This is accomplished in two different ways: (i) chemical engineering of PS to enhance their uptake, e.g. Si-phthalocyanines; and (ii) transgenic approach to render Leishmania inducible for porphyrinogenesis. Three different schemes of Leishmania-based PDV are presented diagrammatically to depict the cellular events resulting in cell-mediated immunity, as seen experimentally against leishmaniasis and Leishmania-delivered antigen in vitro and in vivo. Safety versus efficacy evaluations are under way for PDT-inactivated Leishmania, including those further processed to facilitate their storage and transport. Leishmania transfected to express cancer and viral vaccine candidates are being prepared accordingly for experimental trials. We have begun to examine PS-mediated photodynamic insecticides (PDI). Mosquito cells take up rose bengal/cyanosine, rendering them light-sensitive to undergo disintegration in vitro, thereby providing a cellular basis for the larvicidal activity seen by the same treatments. Ineffectiveness of phthalocyanines and porphyrins for PDI underscores its requirement for different PS. Differential uptake of PS by insect versus other cells to account for this difference is under study. The ongoing work is patterned after the one-world approach by enlisting the participation of experts in medicinal chemistry, cell/molecular biology, immunology, parasitology, entomology, cancer research, tropical medicine and veterinary medicine. The availability of multidisciplinary expertise is indispensable for implementation of the necessary studies to move the project toward product development.
Collapse
Affiliation(s)
- Kwang Poo Chang
- Department of Microbiology/Immunology, Chicago Medical School/Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA.
| | - Bala K Kolli
- Department of Microbiology/Immunology, Chicago Medical School/Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA
| | | |
Collapse
|
15
|
Hou S, Xian L, Shi P, Li C, Lin Z, Gao X. The Magea gene cluster regulates male germ cell apoptosis without affecting the fertility in mice. Sci Rep 2016; 6:26735. [PMID: 27226137 PMCID: PMC4880894 DOI: 10.1038/srep26735] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 05/06/2016] [Indexed: 11/21/2022] Open
Abstract
While apoptosis is essential for male germ cell development, improper activation of apoptosis in the testis can affect spermatogenesis and cause reproduction defects. Members of the MAGE-A (melanoma antigen family A) gene family are frequently clustered in mammalian genomes and are exclusively expressed in the testes of normal animals but abnormally activated in a wide variety of cancers. We investigated the potential roles of these genes in spermatogenesis by generating a mouse model with a 210-kb genomic deletion encompassing six members of the Magea gene cluster (Magea1, Magea2, Magea3, Magea5, Magea6 and Magea8). Male mice carrying the deletion displayed smaller testes from 2 months old with a marked increase in apoptotic germ cells in the first wave of spermatogenesis. Furthermore, we found that Magea genes prevented stress-induced spermatogenic apoptosis after N-ethyl-N-nitrosourea (ENU) treatment during the adult stage. Mechanistically, deletion of the Magea gene cluster resulted in a dramatic increase in apoptotic germ cells, predominantly spermatocytes, with activation of p53 and induction of Bax in the testes. These observations demonstrate that the Magea genes are crucial in maintaining normal testicular size and protecting germ cells from excessive apoptosis under genotoxic stress.
Collapse
Affiliation(s)
- Siyuan Hou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, China
| | - Li Xian
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, China
| | - Peiliang Shi
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, China
| | - Chaojun Li
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, China
| | - Zhaoyu Lin
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, China
| | - Xiang Gao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, China
| |
Collapse
|
16
|
Sun L, Guo H, Jiang R, Lu L, Liu T, He X. Engineered cytotoxic T lymphocytes with AFP-specific TCR gene for adoptive immunotherapy in hepatocellular carcinoma. Tumour Biol 2016; 37:799-806. [PMID: 26250457 DOI: 10.1007/s13277-015-3845-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 07/24/2015] [Indexed: 12/12/2022] Open
Abstract
Alpha-fetoprotein (AFP) is overexpressed in hepatocellular carcinoma (HCC) and could serve as a tumor-associated antigen (TAA) and potential target for adoptive immunotherapy. However, low frequency and severe functional impairment of AFP-specific T cells in vivo hamper adoptive infusion. TAA-specific T cell receptor (TCR) gene transfer could be an efficient and reliable alternation to generate AFP-specific cytotoxic T lymphocytes (CTLs). Autologous dendritic cells (DC) pulsed with AFP158-166 peptides were used to stimulate AFP-specific CTLs. TCR α/β chain genes of AFP-specific CTLs were cloned and linked by 2A peptide to form full-length TCR coding sequence synthesized into a lentiviral vector. Nonspecific activated T cells were engineered by lentivirus infection. Transgenetic CTLs were evaluated for transfection efficiency, expression of AFP158-166-specific TCR, interferon (IFN)-γ secretion, and specific cytotoxicity toward AFP+ HCC cells in vitro and in vivo. Flow cytometry revealed the AFP158-166-MHC-Pentamer positive transgenetic CTLs was 9.86 %. The number of IFN-γ secretion T cells and the specific cytotoxicity toward HpeG2 in vitro and in tumor-bearing NOD/SCID mice were significantly raised in transgenetic CTLs than that of AFP158-166-specific CTLs obtained by peptide-pulsed DCs or control group. TCR gene transfer is a promising strategy to generate AFP158-166-specific CTLs for the treatment of HCC.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/metabolism
- Cell Line, Tumor
- Cytotoxicity, Immunologic
- Enzyme-Linked Immunosorbent Assay
- Female
- Gene Expression Regulation, Neoplastic
- Gene Transfer Techniques
- Genes, T-Cell Receptor
- Hep G2 Cells
- Humans
- Immunotherapy, Adoptive/methods
- Interferon-gamma/metabolism
- L-Lactate Dehydrogenase/metabolism
- Liver Neoplasms/metabolism
- MCF-7 Cells
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Monocytes/cytology
- Peptides/chemistry
- Receptors, Antigen, T-Cell/immunology
- T-Lymphocytes, Cytotoxic/cytology
- Transgenes
- alpha-Fetoproteins/metabolism
Collapse
Affiliation(s)
- Longhao Sun
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road Heping District, Tianjin, 300052, China
| | - Hao Guo
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road Heping District, Tianjin, 300052, China
| | - Ruoyu Jiang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road Heping District, Tianjin, 300052, China
| | - Li Lu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road Heping District, Tianjin, 300052, China
| | - Tong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road Heping District, Tianjin, 300052, China
| | - Xianghui He
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road Heping District, Tianjin, 300052, China.
| |
Collapse
|