1
|
Perri F, Sabbatino F, Ottaiano A, Fusco R, Caraglia M, Cascella M, Longo F, Rega RA, Salzano G, Pontone M, Marciano ML, Piccirillo A, Montano M, Fasano M, Ciardiello F, Della Vittoria Scarpati G, Ionna F. Impact of Epstein Barr Virus Infection on Treatment Opportunities in Patients with Nasopharyngeal Cancer. Cancers (Basel) 2023; 15:1626. [PMID: 36900413 PMCID: PMC10000842 DOI: 10.3390/cancers15051626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
Chemical, physical, and infectious agents may induce carcinogenesis, and in the latter case, viruses are involved in most cases. The occurrence of virus-induced carcinogenesis is a complex process caused by an interaction across multiple genes, mainly depending by the type of the virus. Molecular mechanisms at the basis of viral carcinogenesis, mainly suggest the involvement of a dysregulation of the cell cycle. Among the virus-inducing carcinogenesis, Epstein Barr Virus (EBV) plays a major role in the development of both hematological and oncological malignancies and importantly, several lines of evidence demonstrated that nasopharyngeal carcinoma (NPC) is consistently associated with EBV infection. Cancerogenesis in NPC may be induced by the activation of different EBV "oncoproteins" which are produced during the so called "latency phase" of EBV in the host cells. Moreover, EBV presence in NPC does affect the tumor microenvironment (TME) leading to a strongly immunosuppressed status. Translational implications of the above-mentioned statements are that EBV-infected NPC cells can express proteins potentially recognized by immune cells in order to elicit a host immune response (tumor associated antigens). Three immunotherapeutic approaches have been implemented for the treatment of NPC including active, adoptive immunotherapy, and modulation of immune regulatory molecules by use of the so-called checkpoint inhibitors. In this review, we will highlight the role of EBV infection in NPC development and analyze its possible implications on therapy strategies.
Collapse
Affiliation(s)
- Francesco Perri
- Medical and Experimental Head and Neck Oncology Unit, INT IRCCS Foundation G. Pascale, 80131 Napoli, Italy
| | | | - Alessandro Ottaiano
- SSD Innovative Therapies for Abdominal Metastases, Department of Abdominal Oncology, INT IRCCS Foundation G. Pascale, 80131 Napoli, Italy
| | - Roberta Fusco
- Medical Oncology Division, IGEA SPA, 41012 Naples, Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy
| | - Marco Cascella
- Division of Anesthesia and Pain Medicine, INT IRCCS Foundation G Pascale, 80131 Napoli, Italy
| | - Francesco Longo
- Otolaryngology and Maxillofacial Surgery Surgery Unit, INT IRCCS Foundation G Pascale, 80131 Napoli, Italy
| | - Rosalia Anna Rega
- Medical and Experimental Head and Neck Oncology Unit, INT IRCCS Foundation G. Pascale, 80131 Napoli, Italy
| | - Giovanni Salzano
- Maxillofacial Surgery Surgery Unit, Reproductive and Odontostomatological Science, University of Naples Federico II, 80138 Napoli, Italy
| | - Monica Pontone
- Medical and Experimental Head and Neck Oncology Unit, INT IRCCS Foundation G. Pascale, 80131 Napoli, Italy
| | - Maria Luisa Marciano
- Medical and Experimental Head and Neck Oncology Unit, INT IRCCS Foundation G. Pascale, 80131 Napoli, Italy
| | - Arianna Piccirillo
- Medical and Experimental Head and Neck Oncology Unit, INT IRCCS Foundation G. Pascale, 80131 Napoli, Italy
| | - Massimo Montano
- Medical and Experimental Head and Neck Oncology Unit, INT IRCCS Foundation G. Pascale, 80131 Napoli, Italy
| | - Morena Fasano
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy
| | | | - Franco Ionna
- Division of Anesthesia and Pain Medicine, INT IRCCS Foundation G Pascale, 80131 Napoli, Italy
| |
Collapse
|
2
|
Huang J, Harris E, Lorch J. Vaccination as a therapeutic strategy for Nasopharyngeal carcinoma. Oral Oncol 2022; 135:106083. [DOI: 10.1016/j.oraloncology.2022.106083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/01/2022] [Accepted: 08/10/2022] [Indexed: 11/06/2022]
|
3
|
Jiromaru R, Nakagawa T, Yasumatsu R. Advanced Nasopharyngeal Carcinoma: Current and Emerging Treatment Options. Cancer Manag Res 2022; 14:2681-2689. [PMID: 36117730 PMCID: PMC9480178 DOI: 10.2147/cmar.s341472] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/07/2022] [Indexed: 12/08/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) has seen improved treatment outcomes and a decrease in incidence worldwide in recent years due to developments in medicine and improved public health. However, 70% of cases are still diagnosed at advanced stages and these advanced NPC cases show a poor prognosis. Reports on current and future treatment in advanced NPC are summarized. Chemoradiotherapy is the mainstay of treatment for advanced NPC. The administration of platinum agents as a concurrent drug and intensity modulated radiotherapy (IMRT) is the most appropriate irradiation method, and is associated with high local control rates. For induction and adjuvant chemotherapy, platinum-based two- or three-drug combination chemotherapy is recommended. The tumour volume, plasma Epstein-Barr virus (EBV)-DNA levels, and the tumour site are used to determine the indication for adjuvant and neo-adjuvant chemotherapy. The tolerability of induction chemotherapy is controversial, and the indications and timing should be carefully considered in each case. Chemotherapy is used for patients with distant metastasis. Gemcitabine/cisplatin is the first-line regimen. The efficacy of immune checkpoint inhibitor (ICI) treatment has recently been reported for NPC and, as in other areas of the head and neck, it is expected to be effective for patients with recurrent/distant metastasis. Trials are underway for various uses of ICIs, including induction chemotherapy, postoperative treatment, and use in combination with chemoradiotherapy. Immunotherapy for NPC, an EBV-associated cancer, has been reported to have some efficacy with immunotherapy used in other EBV-associated cancers. Immunotherapy may be introduced for NPC in the future, depending on the results of clinical trials. Future changes in the treatment of NPC are expected to include risk classification based on plasma EBV-DNA levels and the development of personalized treatment with individual selection of timing and type of therapy.
Collapse
Affiliation(s)
- Rina Jiromaru
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Nakagawa
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryuji Yasumatsu
- Department of Otorhinolaryngology Head and Neck Surgery, Faculty of Medicine, Kindai University, Osaka, Japan
| |
Collapse
|
4
|
Lo AKF, Dawson CW, Lung HL, Wong KL, Young LS. The Role of EBV-Encoded LMP1 in the NPC Tumor Microenvironment: From Function to Therapy. Front Oncol 2021; 11:640207. [PMID: 33718235 PMCID: PMC7947715 DOI: 10.3389/fonc.2021.640207] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is closely associated with Epstein-Barr virus (EBV) infection. It is also characterized by heavy infiltration with non-malignant leucocytes. The EBV-encoded latent membrane protein 1 (LMP1) is believed to play an important role in NPC pathogenesis by virtue of its ability to activate multiple cell signaling pathways which collectively promote cell proliferation and survival, angiogenesis, invasiveness, and aerobic glycolysis. LMP1 also affects cell-cell interactions, antigen presentation, and cytokine and chemokine production. Here, we discuss how LMP1 modulates local immune responses that contribute to the establishment of the NPC tumor microenvironment. We also discuss strategies for targeting the LMP1 protein as a novel therapy for EBV-driven malignancies.
Collapse
Affiliation(s)
| | | | - Hong Lok Lung
- Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Ka-Leung Wong
- Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Lawrence S. Young
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
5
|
Cancer Vaccines: Antigen Selection Strategy. Vaccines (Basel) 2021; 9:vaccines9020085. [PMID: 33503926 PMCID: PMC7911511 DOI: 10.3390/vaccines9020085] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Unlike traditional cancer therapies, cancer vaccines (CVs) harness a high specificity of the host’s immunity to kill tumor cells. CVs can train and bolster the patient’s immune system to recognize and eliminate malignant cells by enhancing immune cells’ identification of antigens expressed on cancer cells. Various features of antigens like immunogenicity and avidity influence the efficacy of CVs. Therefore, the choice and application of antigens play a critical role in establishing and developing CVs. Tumor-associated antigens (TAAs), a group of proteins expressed at elevated levels in tumor cells but lower levels in healthy normal cells, have been well-studied and developed in CVs. However, immunological tolerance, HLA restriction, and adverse events are major obstacles that threaten TAA-based CVs’ efficacy due to the “self-protein” characteristic of TAAs. As “abnormal proteins” that are completely absent from normal cells, tumor-specific antigens (TSAs) can trigger a robust immune response against tumor cells with high specificity and without going through central tolerance, contributing to cancer vaccine development feasibility. In this review, we focus on the unique features of TAAs and TSAs and their application in vaccines, summarizing their performance in preclinical and clinical trials.
Collapse
|
6
|
Yan Y, Xue QJ, Liu A, Wang H, Zhang H, Wang S, Zhao L, Li Y, Li X, Yang Y, Chen T, Li S. EB virus-positive tumors are inhibited by rBCG expressing hGM-CSF and LMP2A. Hum Vaccin Immunother 2020; 16:654-663. [PMID: 31567046 DOI: 10.1080/21645515.2019.1670593] [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] [Indexed: 10/25/2022] Open
Abstract
For the development of safe and effective EBV (Epstein-Barr virus) vaccines, the Ag85A signal peptide from M. tuberculosis H37Rv was used to construct a recombinant secretory BCG (Bacillus Chalmette-Guérin) plasmid. The Ag85A gene, fused to the EBV LMP2A (latent membrane protein) and hGM-CSF (human granulocyte/macrophage colony-stimulating factor) genes, was inserted into the pMV261 vector (secretory BCG plasmid). The expression levels of the hGM-CSF and LMP2A proteins in rBCG (recombinant BCG) were measured by Western blot analysis. Humoral immunity, cellular immunity, and antitumor effects were determined by a series of experiments. The recombinant pMVGCA plasmid effectively expressed GCA (hGM-CSF and LMP2A fusion protein) in BCG after transformation, and the rBCG proteins were recognized by antibodies against hGM-CSF and LMP2A. Six weeks after immunization, the maximum dose of rBCG resulted in antibody titers of 1:19,800 (hGM-CSF antibody) and 1:21,800 (LMP2A antibody). When the effector:target ratio was 40:1, specific lysis was maximal and approximately two times stronger than that in mice immunized with the control. Tumorigenicity was lower in the rBCG treatment group, with a tumor inhibition rate of 0.81 ± 0.09 compared with the control groups. EB virus-positive tumors are inhibited by rBCG expressing an hGM-CSF and LMP2A fusion protein.
Collapse
Affiliation(s)
- Yingchun Yan
- School of Mental Health, Jining Medical University, Shandong, China
| | - Qing-Jie Xue
- School of Basic Medical, Jining Medical University, Shandong, China
| | - Ang Liu
- School of Basic Medical, Jining Medical University, Shandong, China
| | - Hui Wang
- School of Basic Medical, Jining Medical University, Shandong, China
| | - Honghua Zhang
- School of Basic Medical, Jining Medical University, Shandong, China
| | - Shuang Wang
- School of Basic Medical, Jining Medical University, Shandong, China
| | - Longyu Zhao
- School of Basic Medical, Jining Medical University, Shandong, China
| | - Yunqing Li
- School of Basic Medical, Jining Medical University, Shandong, China
| | - Xiuzhen Li
- School of Basic Medical, Jining Medical University, Shandong, China
| | - Yuanyuan Yang
- School of Basic Medical, Jining Medical University, Shandong, China
| | - Ting Chen
- School of Basic Medical, Jining Medical University, Shandong, China
| | - Shigen Li
- School of Basic Medical, Jining Medical University, Shandong, China
| |
Collapse
|
7
|
Anti-tumour research of recombinant BCG using BZLF1 and hGM-CSF fusion genes. Vaccine 2017; 35:1599-1607. [DOI: 10.1016/j.vaccine.2017.02.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 12/29/2016] [Accepted: 02/09/2017] [Indexed: 12/12/2022]
|
8
|
Si Y, Deng Z, Lan G, Du H, Wang Y, Si J, Wei J, Weng J, Qin Y, Huang B, Yang Y, Qin Y. The Safety and Immunological Effects of rAd5-EBV-LMP2 Vaccine in Nasopharyngeal Carcinoma Patients: A Phase I Clinical Trial and Two-Year Follow-Up. Chem Pharm Bull (Tokyo) 2017; 64:1118-23. [PMID: 27477649 DOI: 10.1248/cpb.c16-00114] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Epstein-Barr virus (EBV)-encoded latent membrane protein 2 (LMP2) promotes nasopharyngeal carcinoma (NPC) progression. Previously, we reported that the dendritic cells (DCs) transfected with EBV-LMP2 recombinant serotype 5 adenoviruses (rAd5) induced anti-tumor effect by eliciting cytotoxic T lymphocytes (CTLs)-mediated immune response in vitro and the adenoviral vaccine of EBV-LMP2 (rAd5-EBV-LMP2) stimulated antigen-specific cellular immunity in mice. However, the safety and immunological effect of rAd5-EBV-LMP2 vaccine in human still remained unknown. Here we conducted a single-center, non-randomized, open-label, single-arm phase I clinical trial to clarify this unsolved issue. A total of 24 patients with regional advanced NPC were sequentially enrolled into three dose level groups (2×10(9), 2×10(10), 2×10(11) vp). The rAd5-EBV-LMP2 vaccines were intramuscularly injected for four times within 28 d (D0, D7, D14, D28). Blood samples were harvested immediately before every vaccination, one week and one month after the last vaccination (D0, D7, D14, D28, D35, D58). All the vaccine inoculation-related toxicities presented as grade I/II adverse events. The most frequent systemic adverse reactions were fatigue (33.0%, 8/24), myalgia (29.2%, 7/24) and cough (29.2%, 7/24), while the most common regional adverse reaction was tenderness in the inoculation site (54.2%, 13/24). In addition, proportion of CD(3+)CD(4+) cells in peripheral blood was significantly increased in the high dose group (2×10(11) vp). The rAd5-EBV-LMP2 vaccine was generally well-tolerated and the high dose (2×10(11) vp) is recommended to be adopted in phase II studies. The long-term outcome of rAd5-EBV-LMP2 vaccine inoculation is required to be determined in following placebo-controlled trials.
Collapse
Affiliation(s)
- Yongfeng Si
- Department of Otolaryngology-Head and Neck Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Smith C, Lee V, Schuessler A, Beagley L, Rehan S, Tsang J, Li V, Tiu R, Smith D, Neller MA, Matthews KK, Gostick E, Price DA, Burrows J, Boyle GM, Chua D, Panizza B, Porceddu SV, Nicholls J, Kwong D, Khanna R. Pre-emptive and therapeutic adoptive immunotherapy for nasopharyngeal carcinoma: Phenotype and effector function of T cells impact on clinical response. Oncoimmunology 2017; 6:e1273311. [PMID: 28344888 PMCID: PMC5353921 DOI: 10.1080/2162402x.2016.1273311] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/07/2016] [Accepted: 12/13/2016] [Indexed: 01/08/2023] Open
Abstract
Adoptive T cell therapy has emerged as a powerful strategy to treat human cancers especially haematological malignancies. Extension of these therapies to solid cancers remains a significant challenge especially in the context of defining immunological correlates of clinical responses. Here we describe results from a clinical study investigating autologous Epstein-Barr virus (EBV)-specific T cells generated using a novel AdE1-LMPpoly vector to treat patients with nasopharyngeal carcinoma (NPC) either pre-emptively in at-risk patients with no or minimal residual disease (N/MRD) or therapeutically in patients with active recurrent/metastatic disease (ARMD). Tolerability, safety and efficacy, including progression-free survival (PFS) and overall survival (OS), were evaluated following adoptive T-cell immunotherapy. Twenty-nine patients, including 20 with ARMD and nine with N/MRD, successfully completed T-cell therapy. After a median follow-up of 18.5 months, the median PFS was 5.5 months (95% CI 2.1 to 9.0 months) and the median OS was 38.1 months (95% CI 17.2 months to not reached). Post-immunotherapy analyses revealed that disease stabilization in ARMD patients was significantly associated with the functional and phenotypic composition of in vitro-expanded T cell immunotherapy. These included a higher proportion of effector CD8+ T-cells and an increased number of EBV-specific T-cells with broader antigen specificity. These observations indicate that adoptive immunotherapy with AdE1-LMPpoly-expanded T cells stabilizes relapsed, refractory NPC without significant toxicity. Promising clinical outcomes in N/MRD patients further suggest a potential role for this approach as a consolidation treatment following first-line chemotherapy.
Collapse
Affiliation(s)
- Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute , Brisbane, Queensland, Australia
| | - Victor Lee
- Department of Clinical Oncology, Queen Mary Hospital, The University of Hong Kong , Hong Kong
| | - Andrea Schuessler
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute , Brisbane, Queensland, Australia
| | - Leone Beagley
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute , Brisbane, Queensland, Australia
| | - Sweera Rehan
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute , Brisbane, Queensland, Australia
| | - Janice Tsang
- Department of Clinical Oncology, Queen Mary Hospital, The University of Hong Kong , Hong Kong
| | - Vivian Li
- Department of Clinical Oncology, Queen Mary Hospital, The University of Hong Kong , Hong Kong
| | - Randal Tiu
- Department of Clinical Oncology, Queen Mary Hospital, The University of Hong Kong , Hong Kong
| | - David Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute , Brisbane, Queensland, Australia
| | - Michelle A Neller
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute , Brisbane, Queensland, Australia
| | - Katherine K Matthews
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute , Brisbane, Queensland, Australia
| | - Emma Gostick
- Institute of Infection and Immunity, Cardiff University School of Medicine , Cardiff, UK
| | - David A Price
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK; Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jacqueline Burrows
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute , Brisbane, Queensland, Australia
| | - Glen M Boyle
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute , Brisbane, Queensland, Australia
| | - Daniel Chua
- Comprehensive Oncology Centre, Hong Kong Sanatorium Hospital , Hong Kong
| | - Benedict Panizza
- Department of Otolaryngology-Head and Neck Surgery, The Princess Alexandra Hospital, University of Queensland , Brisbane, Queensland, Australia
| | - Sandro V Porceddu
- Department of Otolaryngology-Head and Neck Surgery, The Princess Alexandra Hospital, University of Queensland , Brisbane, Queensland, Australia
| | - John Nicholls
- Department of Pathology, Queen Mary Hospital, The University of Hong Kong , Hong Kong
| | - Dora Kwong
- Department of Clinical Oncology, Queen Mary Hospital, The University of Hong Kong , Hong Kong
| | - Rajiv Khanna
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute , Brisbane, Queensland, Australia
| |
Collapse
|
10
|
Hartlage AS, Liu T, Patton JT, Garman SL, Zhang X, Kurt H, Lozanski G, Lustberg ME, Caligiuri MA, Baiocchi RA. The Epstein-Barr Virus Lytic Protein BZLF1 as a Candidate Target Antigen for Vaccine Development. Cancer Immunol Res 2015; 3:787-94. [PMID: 25735952 DOI: 10.1158/2326-6066.cir-14-0242] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/17/2015] [Indexed: 01/15/2023]
Abstract
The Epstein-Barr virus (EBV) is an oncogenic, γ-herpesvirus associated with a broad spectrum of disease. Although most immune-competent individuals can effectivley develop efficient adaptive immune responses to EBV, immunocompromised individuals are at serious risk for developing life-threatening diseases, such as Hodgkin lymphoma and posttransplant lymphoproliferative disorder (PTLD). Given the significant morbidity associated with EBV infection in high-risk populations, there is a need to develop vaccine strategies that restore or enhance EBV-specific immune responses. Here, we identify the EBV immediate-early protein BZLF1 as a potential target antigen for vaccine development. Primary tumors from patients with PTLD and a chimeric human-murine model of EBV-driven lymphoproliferative disorder (EBV-LPD) express BZLF1 protein. Pulsing human dendritic cells (DC) with recombinant BZLF1 followed by incubation with autologous mononuclear cells led to expansion of BZLF1-specific CD8(+) T cells in vitro and primed BZLF1-specific T-cell responses in vivo. In addition, vaccination of hu-PBL-SCID mice with BZLF1-transduced DCs induced specific cellular immunity and significantly prolonged survival from fatal EBV-LPD. These findings identify BZLF1 as a candidate target protein in the immunosurveillance of EBV and provide a rationale for considering BZLF1 in vaccine strategies to enhance primary and recall immune responses and potentially prevent EBV-associated diseases.
Collapse
Affiliation(s)
- Alex S Hartlage
- The Comprehensive Cancer Center and Arthur G. James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, Ohio
| | - Tom Liu
- The Comprehensive Cancer Center and Arthur G. James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, Ohio
| | - John T Patton
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Sabrina L Garman
- The Comprehensive Cancer Center and Arthur G. James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, Ohio
| | - Xiaoli Zhang
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - Habibe Kurt
- Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Gerard Lozanski
- Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Mark E Lustberg
- Division of Infectious Disease, The Ohio State University, Columbus, Ohio
| | - Michael A Caligiuri
- The Comprehensive Cancer Center and Arthur G. James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, Ohio. Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio.
| | - Robert A Baiocchi
- The Comprehensive Cancer Center and Arthur G. James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, Ohio. Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
11
|
Shen Y, Zhang S, Sun R, Wu T, Qian J. Understanding the interplay between host immunity and Epstein-Barr virus in NPC patients. Emerg Microbes Infect 2015; 4:e20. [PMID: 26038769 PMCID: PMC4395660 DOI: 10.1038/emi.2015.20] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 12/22/2014] [Accepted: 02/23/2015] [Indexed: 12/23/2022]
Abstract
Epstein-Barr virus (EBV) has been used as a paradigm for studying host-virus interactions, not only because of its importance as a human oncogenic virus associated with several malignancies including nasopharyngeal carcinoma (NPC) but also owing to its sophisticated strategies to subvert the host antiviral responses. An understanding of the interplay between EBV and NPC is critical for the development of EBV-targeted immunotherapy. Here, we summarize the current knowledge regarding the host immune responses and EBV immune evasion mechanisms in the context of NPC.
Collapse
Affiliation(s)
- Yong Shen
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, Cancer Institute, Zhejiang University School of Medicine , Hangzhou 310009, Zhejiang Province, China ; ZJU-UCLA Joint Center for Medical Education and Research, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine , Hangzhou 310058, Zhejiang Province, China
| | - Suzhan Zhang
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, Cancer Institute, Zhejiang University School of Medicine , Hangzhou 310009, Zhejiang Province, China ; ZJU-UCLA Joint Center for Medical Education and Research, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine , Hangzhou 310058, Zhejiang Province, China
| | - Ren Sun
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, Cancer Institute, Zhejiang University School of Medicine , Hangzhou 310009, Zhejiang Province, China ; ZJU-UCLA Joint Center for Medical Education and Research, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine , Hangzhou 310058, Zhejiang Province, China ; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles , Los Angeles, California 90095, USA
| | - Tingting Wu
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, Cancer Institute, Zhejiang University School of Medicine , Hangzhou 310009, Zhejiang Province, China ; ZJU-UCLA Joint Center for Medical Education and Research, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine , Hangzhou 310058, Zhejiang Province, China ; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles , Los Angeles, California 90095, USA
| | - Jing Qian
- ZJU-UCLA Joint Center for Medical Education and Research, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine , Hangzhou 310058, Zhejiang Province, China ; Research Center of Infection and Immunity, Zhejiang University School of Medicine , Hangzhou 310058, Zhejiang Province, China
| |
Collapse
|
12
|
Xue SA, Gao L, Ahmadi M, Ghorashian S, Barros RD, Pospori C, Holler A, Wright G, Thomas S, Topp M, Morris EC, Stauss HJ. Human MHC Class I-restricted high avidity CD4 + T cells generated by co-transfer of TCR and CD8 mediate efficient tumor rejection in vivo. Oncoimmunology 2014; 2:e22590. [PMID: 23483821 PMCID: PMC3583927 DOI: 10.4161/onci.22590] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In this study, we generated human MHC Class I-restricted CD4+ T cells specific for Epstein-Barr virus (EBV) and cytomegalovirus (CMV), two herpesviridae associated with lymphoma, nasopharyngeal carcinoma and medulloblastoma, respectively. Retroviral transfer of virus-specific, HLA-A2-restricted TCR-coding genes generated CD4+ T cells that recognized HLA-A2/peptide multimers and produced cytokines when stimulated with MHC Class II-deficient cells presenting the relevant viral peptides in the context of HLA-A2. Peptide titration revealed that CD4+ T cells had a 10-fold lower avidity than CD8+ T cells expressing the same TCR. The impaired avidity of CD4+ T cells was corrected by simultaneously transferring TCR- and CD8-coding genes. The CD8 co-receptor did not alter the cytokine signature of CD4+ T cells, which remained distinct from that of CD8+ T cells. Using the xenogeneic NOD/SCID mouse model, we demonstrated that human CD4+ T cells expressing a specific TCR and CD8 can confer efficient protection against the growth of tumors expressing the EBV or CMV antigens recognized by the TCR. In summary, we describe a robust approach for generating therapeutic CD4+ T cells capable of providing MHC Class I-restricted immunity against MHC Class II-negative tumors in vivo.
Collapse
Affiliation(s)
- Shao-An Xue
- Department of Immunology; University College London; Royal Free Hospital; London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Hutajulu SH, Kurnianda J, Tan IB, Middeldorp JM. Therapeutic implications of Epstein-Barr virus infection for the treatment of nasopharyngeal carcinoma. Ther Clin Risk Manag 2014; 10:721-36. [PMID: 25228810 PMCID: PMC4161530 DOI: 10.2147/tcrm.s47434] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is highly endemic in certain regions including the People’s Republic of China and Southeast Asia. Its etiology is unique and multifactorial, involving genetic background, epigenetic, and environment factors, including Epstein–Barr virus (EBV) infection. The presence of EBV in all tumor cells, aberrant pattern of antibodies against EBV antigens in patient sera, and elevated viral DNA in patient circulation as well as nasopharyngeal site underline the role of EBV during NPC development. In NPC tumors, EBV expresses latency type II, where three EBV-encoded proteins, Epstein–Barr nuclear antigen 1, latent membrane protein 1 and 2 (LMP1, 2), are expressed along with BamH1-A rightward reading frame 1, Epstein–Barr virus-encoded small nuclear RNAs, and BamH1-A rightward transcripts. Among all encoded proteins, LMP1 plays a central role in the propagation of NPC. Standard treatment of NPC consists of radiotherapy with or without chemotherapy for early stage, concurrent chemoradiotherapy in locally advanced tumors, and palliative systemic chemotherapy in metastatic disease. However, this standard care has limitations, allowing recurrences and disease progression in a certain proportion of cases. Although the pathophysiological link and molecular process of EBV-induced oncogenesis are not fully understood, therapeutic approaches targeting the virus may increase the cure rate and add clinical benefit. The promising results of early phase clinical trials on EBV-specific immunotherapy, epigenetic therapy, and treatment with viral lytic induction offer new options for treating NPC.
Collapse
Affiliation(s)
- Susanna Hilda Hutajulu
- Department of Internal Medicine, Faculty of Medicine Universitas Gadjah Mada/Dr Sardjito General Hospital, Yogyakarta, Indonesia
| | - Johan Kurnianda
- Department of Internal Medicine, Faculty of Medicine Universitas Gadjah Mada/Dr Sardjito General Hospital, Yogyakarta, Indonesia
| | - I Bing Tan
- Department of Ear, Nose and Throat, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands ; Department of Ear, Nose and Throat, Faculty of Medicine Universitas Gadjah Mada/Dr Sardjito General Hospital, Yogyakarta, Indonesia
| | - Jaap M Middeldorp
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Xue QJ, Dai J, Li XZ, Zhu W, Si CP, Chen T. Construction of a recombinant-BCG containing the LMP2A and BZLF1 genes and its significance in the Epstein-Barr virus positive gastric carcinoma. J Med Virol 2014; 86:1780-7. [PMID: 24699993 DOI: 10.1002/jmv.23901] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2014] [Indexed: 01/28/2023]
Abstract
The signal peptide Ag85B of Bacillus Chalmette-Guerin (BCG) was used to construct a recombinant plasmid of BCG. The BCG-Ag85B gene and fused EBV LMP2A and BZLF1 genes were amplified and successively inserted into the Escherichia coli-BCG shuttle-vector pMV261. The recombinant plasmids were then amplified in E. coli DH5α and transformed into competent BCG. The expression of BZLF1 and LMP2A fusion proteins in recombinant-BCG (rBCG) was shown by Western blot. After the injection of recombinant-BCG into mice, antibodies against the fusion protein BZLF1 and LMP2A were measured by ELISA, and the cellular immune effects were determined by the lactate dehydrogenate (LDH) release assays. The results confirmed that the cloned genes of BCG-Ag85B and Z2A were correctly inserted into the vector pMV261. The recombinant plasmid pMVZ2A expressed Z2A in BCG effectively after transformation. The rBCG proteins were recognized by the BZLF1 (LMP2A) antibody. An ELISA demonstrated that rBCG could stimulate the generation of antibody against the fusion protein. The fusion gene was constructed successfully, and the rBCG induced humoral and cellular immune response in mice.
Collapse
Affiliation(s)
- Qing-Jie Xue
- Department of Pathogenic Biology, Provincial Key Discipline of Medical Immunology, Jining Medical University, Shandong, China
| | | | | | | | | | | |
Collapse
|
15
|
Tsang J, Lee VHF, Kwong DLW. Novel therapy for nasopharyngeal carcinoma--where are we. Oral Oncol 2014; 50:798-801. [PMID: 24462373 DOI: 10.1016/j.oraloncology.2014.01.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 01/04/2014] [Indexed: 01/24/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is endemic in Southern China, and the South-East Asia including Hong Kong. We still see patients recur after primary treatment with radiotherapy or chemo-irradiation. Management of nasopharyngeal carcinoma remains one of the biggest clinical challenges. There have been breakthroughs in early detection, diagnosis, multi-modality treatment and also disease monitoring for NPC. Systemic treatment has been crucial to the management of locally advanced or metastatic NPC. With the advent of molecular targeted therapy and personalized medicine, novel therapies based on molecular targets of NPC have become the focus of research and development over the last decade. Furthermore, as NPC is tightly associated with the Epstein-Barr virus (EBV) infection, the role of tumor-associated viral antigens in NPC renders it an appealing candidate for cellular immunotherapy. This is a review of recent evolving concerted efforts and the success from our translational research with focus of the recent systemic novel targeted therapies including the potential role of immunotherapy which may offer further clinical benefit to our patients living with NPC. The scientific basis and latest published results of the relevant clinical trials are highlighted, demonstrating the ongoing battle against NPC is indeed one of the most fascinating successes in head and neck oncology.
Collapse
Affiliation(s)
- Janice Tsang
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Victor H F Lee
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Dora L W Kwong
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
16
|
Epstein-Barr virus vaccines. Vaccines (Basel) 2013. [DOI: 10.1016/b978-1-4557-0090-5.00050-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
17
|
Basic consideration of research strategies for head and neck cancer. Front Med 2012; 6:339-53. [DOI: 10.1007/s11684-012-0213-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 06/18/2012] [Indexed: 10/27/2022]
|
18
|
Lee IS. Epstein-Barr Virus-Associated Classical Hodgkin Lymphoma and Its Therapeutic Strategies. Biomol Ther (Seoul) 2011. [DOI: 10.4062/biomolther.2011.19.4.398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
19
|
|
20
|
West NP, Thomson SA, Triccas JA, Medveczky CJ, Ramshaw IA, Britton WJ. Delivery of a multivalent scrambled antigen vaccine induces broad spectrum immunity and protection against tuberculosis. Vaccine 2011; 29:7759-65. [PMID: 21846485 DOI: 10.1016/j.vaccine.2011.07.109] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 07/21/2011] [Accepted: 07/25/2011] [Indexed: 12/23/2022]
Abstract
The development of effective anti-Tuberculosis (TB) vaccines is an important step towards improved control of TB in high burden countries. Subunit vaccines are advantageous in terms of safety, particularly in the context of high rates of HIV co-infection, but they must contain sufficient Mycobacterium tuberculosis antigens to stimulate immunity in genetically diverse human populations. We have used a novel approach to develop a synthetic scrambled antigen vaccine (TB-SAVINE), comprised of overlapping, recombined peptides from four M. tuberculosis proteins, Ag85B, ESAT-6, PstS3 and Mpt83, each of which is immunogenic and protective against experimental TB. This polyvalent TB-SAVINE construct stimulated CD4 and CD8T cell responses against the individual proteins and M. tuberculosis in C57BL/6 and Balb/c mice, when delivered as DNA, Fowl Pox Virus or Vaccinia Virus vaccines. In addition, the DNA-TBS vaccine induced protective immunity against pulmonary M. tuberculosis infection in C57BL/6 mice. Co-immunization of Balb/c mice with virally expressed TBS and HIV1-SAVINE vaccine stimulated strong T cell responses to both the M. tuberculosis and HIV proteins, indicating no effects of antigenic competition. Further development of this TB-SAVINE vaccine expressing components from multiple M. tuberculosis proteins may prove an effective vaccine candidate against TB, which could potentially form part of a safe, combined preventative strategy together with HIV immunisations.
Collapse
Affiliation(s)
- Nicholas P West
- Mycobacterial Research Program, Centenary Institute, NSW, 2042, Australia.
| | | | | | | | | | | |
Collapse
|
21
|
Adoptive transfer of EBV-specific T cells results in sustained clinical responses in patients with locoregional nasopharyngeal carcinoma. J Immunother 2011; 33:983-90. [PMID: 20948438 DOI: 10.1097/cji.0b013e3181f3cbf4] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Patients with recurrent or refractory Epstein Barr Virus (EBV)-positive nasopharyngeal carcinoma (NPC) continue to have poor outcomes. Our earlier Phase I dose escalation clinical study of 10 NPC patients showed that infusion of EBV-specific cytotoxic T cells (EBV-CTLs) was safe and had antitumor activity. To better define the overall response rate and discover whether disease status, EBV-antigen specificity, and/or in vivo expansion of infused EBV-CTLs predicted outcome, we treated 13 additional NPC patients with EBV-CTLs in a fixed-dose, Phase II component of the study. We assessed toxicity, efficacy, specificity, and expansion of infused CTLs for all 23 recurrent/refractory NPC patients treated on this Phase I/II clinical study. At the time of CTL infusion, 8 relapsed NPC patients were in remission and 15 had active disease. No significant toxicity was observed. Of the relapsed patients treated in their second or subsequent remission, 62% (5/8) remain disease free (at 17 to 75 mo), whereas 48.7% (7/15) of those with active disease had a CR/CRu (33.3%) or PR (15.4%). In contrast to locoregional disease, metastatic disease was associated with an increased risk of disease progression (HR: 3.91, P=0.015) and decreased overall survival (HR: 5.55, P=0.022). Neither the specificity of the infused CTLs for particular EBV antigens nor their measurable in vivo expansion discernibly influenced outcome. In conclusion, treatment of patients with relapsed/refractory EBV-positive NPC with EBV-CTLs is safe and can be associated with significant, long-term clinical benefit, particularly for patients with locoregional disease.
Collapse
|
22
|
Long HM, Taylor GS, Rickinson AB. Immune defence against EBV and EBV-associated disease. Curr Opin Immunol 2011; 23:258-64. [PMID: 21269819 DOI: 10.1016/j.coi.2010.12.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 12/23/2010] [Indexed: 10/18/2022]
Abstract
Epstein-Barr virus (EBV), a B-lymphotropic herpesvirus widespread in the human population and normally contained as an asymptomatic infection by T cell surveillance, nevertheless causes infectious mononucleosis and is strongly linked to several types of human cancer. Here we describe new findings on the range of cellular immune responses induced by EBV infection, on viral strategies to evade those responses and on the links between HLA gene loci and EBV-induced disease. The success of adoptive T cell therapy for EBV-driven post-transplant lymphoproliferative disease is stimulating efforts to target other EBV-associated tumours by immunotherapeutic means, and has reawakened interest in the ultimate intervention strategy, a prophylactic EBV vaccine.
Collapse
Affiliation(s)
- Heather M Long
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | | | |
Collapse
|
23
|
|