1
|
Mai Q, He B, Deng S, Zeng Q, Xu Y, Wang C, Pang Y, Zhang S, Li J, Zeng J, Huang L, Fu Y, Li C, Li T, Xu X, Zhang L. Efficacy of NKG2D CAR-T cells with IL-15/IL-15Rα signaling for treating Epstein-Barr virus-associated lymphoproliferative disorder. Exp Hematol Oncol 2024; 13:85. [PMID: 39160631 PMCID: PMC11334566 DOI: 10.1186/s40164-024-00553-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024] Open
Abstract
Epstein-Barr virus (EBV) related post-transplant lymphoproliferative disorder (EBV-PTLD) is a life-threatening complication after hematopoietic stem cell transplantation (HSCT) or solid organ transplantation (SOT), for which no standard therapeutic means have been developed. Significant increase expression of natural killer group 2 member D ligands (NKG2DLs) was observed on B-lymphoblastoid cells of EBV-PTLD, indicating NKG2DLs as potential therapeutic targets for treatment of EBV-PTLD. In this study, the recombinant constructs of NKG2D CAR and IL-15/IL-15Rα-NKG2D CAR were generated with a retroviral vector and then transduced to human T cells to produce NKG2D CAR-T and IL-15/IL-15Rα-NKG2D CAR-T cells, respectively. B-lymphoblastoid cell lines (B-LCLs) and the xenografted mouse models were established to evaluate the efficacy of these CAR-T cells. IL-15/IL-15Rα-NKG2D CAR-T cells exhibited superior proliferation and antigen-specific cytotoxic effect compared to NKG2D CAR-T, as IL-15/IL-15Rα signaling promoted the expansion of less differentiated central memory T cells (TCM) and increased expression of CD107a and IFN-γ. Moreover, EBV DNA load was dramatically reduced, and 80% B-LCL cells were eliminated by IL-15/IL-15Rα-NKG2D CAR-T cells after co-culturing. In-vivo study confirmed that IL-15/IL-15Rα-NKG2D CAR-T cell therapy significantly enhanced antiviral efficacy in mice, as the serum load of EBV after IL-15/IL-15Rα-NKG2D CAR-T cell infusion was 1500 times lower than the untreated control (P < 0.001). The enhanced efficacy of IL-15/IL-15Rα-NKG2D CAR T cells was probably due to the IL-15/IL-15Rα signaling improved homing and persistence of NKG2D CAR-T cells in vivo, and increased the production of IFN-γ, Perforin, and Granulysin. In conclusion, NKG2D CAR-T cells co-expressing IL-15/IL-15Rα promoted the central memory CAR T cell proliferation and improved the homing and persistence of CAR T cells in vivo, resulting in enhanced anti-tumor and anti-viral effects in treating EBV-PTLD.
Collapse
Affiliation(s)
- Qiusui Mai
- Department of Blood Transfusion, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Bailin He
- Department of Hematology, Nanfang Hospital, Southern Medical Universit, Guangzhou, 510515, China
| | - Shikai Deng
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Qing Zeng
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Yanwen Xu
- Department of Obstetrics, He Xian Memorial Affiliated Hospital of Southern Medical University, Guangzhou, 511402, China
| | - Cong Wang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
- Guangzhou Bai Rui Kang (BRK) Biological Science and Technology Limited Company, Guangzhou, 510555, China
| | - Yunyi Pang
- Department of Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Sheng Zhang
- Shenzhen Bao'an District Central Blood Station, Shenzhen, 518101, China
| | - Jinfeng Li
- Shenzhen Bao'an District Central Blood Station, Shenzhen, 518101, China
| | | | - Liqin Huang
- Shenzhen Blood Center, Shenzhen, 518035, China
| | - Yongshui Fu
- Guangzhou Blood Center, Guangzhou, 510095, China
| | - Chengyao Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China.
- Guangzhou Bai Rui Kang (BRK) Biological Science and Technology Limited Company, Guangzhou, 510555, China.
| | - Tingting Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China.
- Shenzhen Bao'an District Central Blood Station, Shenzhen, 518101, China.
| | - Xiaojun Xu
- Department of Blood Transfusion, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| | - Ling Zhang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Dudaniec K, Westendorf K, Nössner E, Uckert W. Generation of Epstein-Barr Virus Antigen-Specific T Cell Receptors Recognizing Immunodominant Epitopes of LMP1, LMP2A, and EBNA3C for Immunotherapy. Hum Gene Ther 2021; 32:919-935. [PMID: 33798008 DOI: 10.1089/hum.2020.283] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Epstein-Barr virus (EBV) infections in healthy individuals are usually cleared by immune cells, wherein CD8+ T lymphocytes play the most important role. However, in some immunocompromised individuals, EBV infections can lead to the development of cancer in B, T, natural killer (NK) cells and epithelial cells. Most EBV-associated cancers express a limited number of virus-specific antigens such as latent membrane proteins (LMP1 and LMP2) and nuclear proteins (EBNA1, -2, EBNA3A, -B, -C, and EBNA-LP). These antigens represent true tumor-specific antigens and can be considered useful targets for T cell receptor (TCR) gene therapy to treat EBV-associated diseases. We used a TCR isolation platform based on a single major histocompatibility complex class I (MHC I) K562 cell library for the detection, isolation, and re-expression of TCRs targeting immunodominant peptide MHC (pMHC). Mature dendritic cells (mDCs) were pulsed with in vitro-transcribed (ivt) RNA encoding for the selected antigen to stimulate autologous T cells. The procedure allowed the mDCs to select an immunogenic epitope of the antigen for processing and presentation on the cell surface in combination with the most suitable MHC I molecule. We isolated eight EBV-specific TCRs. They recognize various pMHCs of EBV antigens LMP1, LMP2A, and EBNA3C, some of them described previously and some newly identified in this study. The TCR genes were molecularly cloned into retroviral vectors and the resultant TCR-engineered T cells secreted interferon-γ after antigen contact and were able to lyse tumor cells. The EBV-specific TCRs can be used as a basis for the generation of a TCR library, which provides a valuable source of TCRs for the production of EBV-specific T cells to treat EBV-associated diseases in patients with different MHC I types.
Collapse
Affiliation(s)
- Krystyna Dudaniec
- Molecular Cell Biology and Gene Therapy, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Kerstin Westendorf
- Molecular Cell Biology and Gene Therapy, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | | | - Wolfgang Uckert
- Molecular Cell Biology and Gene Therapy, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
3
|
Migliori E, Chang M, Muranski P. Restoring antiviral immunity with adoptive transfer of ex-vivo generated T cells. Curr Opin Hematol 2018; 25:486-493. [PMID: 30281036 DOI: 10.1097/moh.0000000000000461] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Latent viruses such as cytomegalovirus (CMV), Epstein-Barr virus (EBV) and adenovirus (ADV) often reactivate in immunocompromised patients, contributing to poor clinical outcomes. A rapid reconstitution of antiviral responses via adoptive transfer of virus-specific T cells (VSTs) can prevent or eradicate even refractory infections. Here, we evaluate this strategy and the associated methodological, manufacturing and clinical advances. RECENT FINDINGS From the early pioneering but cumbersome efforts to isolate CMV-specific T cell clones, new approaches and techniques have been developed to provide quicker, safer and broader-aimed ex-vivo antigen-specific cells. New manufacturing strategies, such as the use of G-Rex flasks or 'priming' with a library of overlapping viral peptides, allow for culturing greater numbers of cells that could be patient-specific or stored in cell banks for off-the-shelf applications. Rapid isolation of T cells using major histocompatibility complex tetramer or cytokine capture approaches, or genetic reprogramming of cells to target viral antigens can accelerate the generation of potent cellular products. SUMMARY Advances in the ex-vivo generation of VSTs in academic medical centres and as off-the-shelf blood bank-based or commercially produced reagents are likely to result in broader accessibility and possible manufacturing cost reduction of these cell products, and will open new therapeutic prospects for vulnerable and critically ill immunocompromised patients.
Collapse
Affiliation(s)
- Edoardo Migliori
- Columbia Center for Translational Immunology (CCTI), Division of Hematology/Oncology, Columbia University Medical Center, New York, New York, USA
| | | | | |
Collapse
|
4
|
Dharmadhikari B, Nickles E, Harfuddin Z, Ishak NDB, Zeng Q, Bertoletti A, Schwarz H. CD137L dendritic cells induce potent response against cancer-associated viruses and polarize human CD8 + T cells to Tc1 phenotype. Cancer Immunol Immunother 2018; 67:893-905. [PMID: 29508025 PMCID: PMC11028277 DOI: 10.1007/s00262-018-2144-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/27/2018] [Indexed: 12/14/2022]
Abstract
Therapeutic tumor vaccination based on dendritic cells (DC) is safe; however, its efficacy is low. Among the reasons for only a subset of patients benefitting from DC-based immunotherapy is an insufficient potency of in vitro generated classical DCs (cDCs), made by treating monocytes with GM-CSF + IL-4 + maturation factors. Recent studies demonstrated that CD137L (4-1BBL, TNFSF9) signaling differentiates human monocytes to a highly potent novel type of DC (CD137L-DCs) which have an inflammatory phenotype and are closely related to in vivo DCs. Here, we show that CD137L-DCs induce potent CD8+ T-cell responses against Epstein-Barr virus (EBV) and Hepatitis B virus (HBV), and that T cells primed by CD137L-DCs more effectively lyse EBV+ and HBV+ target cells. The chemokine profile of CD137L-DCs identifies them as inflammatory DCs, and they polarize CD8+ T cells to a Tc1 phenotype. Expression of exhaustion markers is reduced on T cells activated by CD137L-DCs. Furthermore, these T cells are metabolically more active and have a higher capacity to utilize glucose. CD137L-induced monocyte to DC differentiation leads to the formation of AIM2 inflammasome, with IL-1beta contributing to CD137L-DCs possessing a stronger T cell activation ability. CD137L-DCs are effective in crosspresentation. PGE2 as a maturation factor is required for enhancing migration of CD137L-DCs but does not significantly reduce their potency. This study shows that CD137L-DCs have a superior ability to activate T cells and to induce potent Tc1 responses against the cancer-causing viruses EBV and HBV which suggest CD137L-DCs as promising candidates for DC-based tumor immunotherapy.
Collapse
Affiliation(s)
- Bhushan Dharmadhikari
- Department of Physiology and Immunology Programme, National University of Singapore (NUS), 2 Medical Dr., Singapore, 117593, Singapore
| | - Emily Nickles
- Department of Physiology and Immunology Programme, National University of Singapore (NUS), 2 Medical Dr., Singapore, 117593, Singapore
| | - Zulkarnain Harfuddin
- Department of Physiology and Immunology Programme, National University of Singapore (NUS), 2 Medical Dr., Singapore, 117593, Singapore
- NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore, 117456, Singapore
| | - Nur Diana Binte Ishak
- Department of Physiology and Immunology Programme, National University of Singapore (NUS), 2 Medical Dr., Singapore, 117593, Singapore
| | - Qun Zeng
- Department of Physiology and Immunology Programme, National University of Singapore (NUS), 2 Medical Dr., Singapore, 117593, Singapore
| | | | - Herbert Schwarz
- Department of Physiology and Immunology Programme, National University of Singapore (NUS), 2 Medical Dr., Singapore, 117593, Singapore.
- NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore, 117456, Singapore.
| |
Collapse
|
5
|
Mancini N, Marrone L, Clementi N, Sautto GA, Clementi M, Burioni R. Adoptive T-cell therapy in the treatment of viral and opportunistic fungal infections. Future Microbiol 2016; 10:665-82. [PMID: 25865200 DOI: 10.2217/fmb.14.122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Viral infections and opportunistic fungal pathogens represent a major menace for immunocompromised patients. Despite the availability of antifungal and antiviral drugs, mortality in these patients remains high, underlining the need of novel therapeutic options based on completely different strategies. This review describes the potential of several T-cell-based therapeutic approaches in the prophylaxis and treatment of infectious diseases with a particular focus on persistent viral infections and opportunistic fungal infections, as these mostly affect immunocompromised patients.
Collapse
Affiliation(s)
- Nicasio Mancini
- Laboratorio di Microbiologia e Virologia, Università 'Vita-Salute' San Raffaele, DIBIT2, via Olgettina 58, 20132, Milan, Italy
| | | | | | | | | | | |
Collapse
|
6
|
Cancer immunotherapy utilizing gene-modified T cells: From the bench to the clinic. Mol Immunol 2015; 67:46-57. [DOI: 10.1016/j.molimm.2014.12.009] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/12/2014] [Accepted: 12/17/2014] [Indexed: 01/02/2023]
|
7
|
Kershaw MH, Westwood JA, Slaney CY, Darcy PK. Clinical application of genetically modified T cells in cancer therapy. Clin Transl Immunology 2014; 3:e16. [PMID: 25505964 PMCID: PMC4232070 DOI: 10.1038/cti.2014.7] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 04/01/2014] [Accepted: 04/01/2014] [Indexed: 02/08/2023] Open
Abstract
Immunotherapies are emerging as highly promising approaches for the treatment of cancer. In these approaches, a variety of materials are used to boost immunity against malignant cells. A key component of many of these approaches is functional tumor-specific T cells, but the existence and activity of sufficient T cells in the immune repertoire is not always the case. Recent methods of generating tumor-specific T cells include the genetic modification of patient lymphocytes with receptors to endow them with tumor specificity. These T cells are then expanded in vitro followed by infusion of the patient in adoptive cell transfer protocols. Genes used to modify T cells include those encoding T-cell receptors and chimeric antigen receptors. In this review, we provide an introduction to the field of genetic engineering of T cells followed by details of their use against cancer in the clinic.
Collapse
Affiliation(s)
- Michael H Kershaw
- Sir Peter MacCallum Cancer Centre, Department of Oncology, University of Melbourne , Melbourne, Victoria, Australia ; Department of Immunology, Monash University , Prahran, Victoria, Australia
| | - Jennifer A Westwood
- Sir Peter MacCallum Cancer Centre, Department of Oncology, University of Melbourne , Melbourne, Victoria, Australia
| | - Clare Y Slaney
- Sir Peter MacCallum Cancer Centre, Department of Oncology, University of Melbourne , Melbourne, Victoria, Australia
| | - Phillip K Darcy
- Sir Peter MacCallum Cancer Centre, Department of Oncology, University of Melbourne , Melbourne, Victoria, Australia ; Department of Immunology, Monash University , Prahran, Victoria, Australia
| |
Collapse
|
8
|
Abstract
T cells have the capacity to eradicate diseased cells, but tumours present considerable challenges that render T cells ineffectual. Cancer cells often make themselves almost 'invisible' to the immune system, and they sculpt a microenvironment that suppresses T cell activity, survival and migration. Genetic engineering of T cells can be used therapeutically to overcome these challenges. T cells can be taken from the blood of cancer patients and then modified with genes encoding receptors that recognize cancer-specific antigens. Additional genes can be used to enable resistance to immunosuppression, to extend survival and to facilitate the penetration of engineered T cells into tumours. Using genetic modification, highly active, self-propagating 'slayers' of cancer cells can be generated.
Collapse
Affiliation(s)
- Michael H Kershaw
- Cancer Immunology Research Program, Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia. michael.kershaw@ petermac.org
| | | | | |
Collapse
|
9
|
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
|
10
|
Evaluation of Epstein-Barr virus latent membrane protein 2 specific T-cell receptors driven by T-cell specific promoters using lentiviral vector. Clin Dev Immunol 2011; 2011:716926. [PMID: 21969838 PMCID: PMC3182378 DOI: 10.1155/2011/716926] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 07/26/2011] [Accepted: 07/26/2011] [Indexed: 02/05/2023]
Abstract
Transduction of latent membrane protein 2 (LMP2)-specific T-cell receptors into activated T lymphocytes may provide a universal, MHC-restricted mean to treat EBV-associated tumors in adoptive immunotherapy. We compared TCR-specific promoters of distinct origin in lentiviral vectors, that is, Vβ6.7, delta, luria, and Vβ5.1 to evaluate TCR gene expression in human primary peripheral blood monocytes and T cell line HSB2. Vectors containing Vβ 6.7 promoter were found to be optimal for expression in PBMCs, and they maintained expression of the transduced TCRs for up to 7 weeks. These cells had the potential to recognize subdominant EBV latency antigens as measured by cytotoxicity and IFN-γ secretion. The nude mice also exhibited significant resistance to the HLA-A2 and LMP2-positive CNE tumor cell challenge after being infused with lentiviral transduced CTLs. In conclusion, LMP2-specific CTLs by lentiviral transduction have the potential use for treatment of EBV-related tumors.
Collapse
|
11
|
Sangiolo D, Leuci V, Gallo S, Aglietta M, Piacibello W. Gene-modified T lymphocytes in the setting of hematopoietic cell transplantation: potential benefits and possible risks. Expert Opin Biol Ther 2011; 11:655-66. [PMID: 21375466 DOI: 10.1517/14712598.2011.565325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Allogeneic hematopoietic cell transplantation (HCT) is a consolidated treatment for several hematologic malignancies. Donor T lymphocytes can mediate a graft versus tumor (GVT) effect and control opportunistic infections but can also cause severe graft versus host disease (GVHD). Gene-transfer strategies are appealing tools to modulate T cell functions when infused after HCT. AREAS COVERED The current and potential future applications of T cell gene-transfer approaches to HCT. This review is not limited to GVHD control but covers the issues of GVT and immune reconstitution. Clinical data are used to discuss more general issues, perspectives and concerns common to gene-modification of T cells. An overview of the results and limitations emerging from clinical trials with herpes simplex virus-thymidine kinase (HSV-TK) engineered lymphocytes is provided. The review provides perspectives on additional gene-transfer strategies, currently at preclinical level or that have just entered clinical trials, to increase the efficacy and safety of HCT. EXPERT OPINION Gene-transfer can positively interfere with T cell functions after HCT. TK-lymphocytes have proven effective in controlling GVHD while retaining an acceptable GVT effect. Strategies exploiting new suicide molecules or engineered T cell receptors (TCRs) should be further explored to address current limitations with TK-lymphocytes and augment the efficacy and specificity of GVT and antiviral activity.
Collapse
Affiliation(s)
- Dario Sangiolo
- IRCC Institute for Cancer Research and Treatment, Laboratory of Cell Therapy, Department of Oncological Sciences, Strada Provinciale 142, Km 3.95, 10060 Candiolo, Turin, Italy.
| | | | | | | | | |
Collapse
|
12
|
|
13
|
Abstract
BACKGROUND Chemotherapy-resistant lymphomas can be cured with allogeneic hematopoietic cell transplantation, demonstrating the susceptibility of these tumors to T cell mediated immune responses. However, high rates of transplant-related morbidity and mortality limit this approach. Efforts have, therefore, been made to develop alternative T cell based therapies, and there is growing evidence that adoptive therapy with T cells targeted to lymphoma-associated antigens may be a safe and effective new method for treating this group of diseases. OBJECTIVE/METHODS We review publications on adoptive therapy with ex vivo expanded T cells targeting viral antigens, as well as genetically modified autologous T cells, as strategies for the treatment of lymphoma, with the goal of providing an overview of these approaches. RESULTS/CONCLUSIONS Epstein-Barr virus specific T cell therapy is an effective and safe method of treating Epstein-Barr virus associated lymphomas; however, most lymphoma subtypes do not express EBV antigens. For these diseases, adoptive immunotherapy with genetically modified T cells expressing chimeric T cell receptors targeting lymphoma-associated antigens such as CD19 and CD20 appears to be a promising alternative. Recent innovations including enhanced co-stimulation, exogenous cytokine administration and use of memory T cells promise to overcome many of the limitations and pitfalls initially encountered with this approach.
Collapse
Affiliation(s)
- Brian G Till
- Research Associate, Acting Instructor, University of Washington, Fred Hutchinson Cancer Research Center, Department of Medicine, Seattle, WA 98109, USA.
| | | |
Collapse
|
14
|
Abstract
Several associations have been described between the frequency of human leukocyte antigen (HLA) class I genes in certain populations and the risk of developing nasopharyngeal carcinoma (NPC). Associations between ethnic background and geographic distribution, and relative disease incidence have been reported. Populations in geographical areas at higher risk of developing NPC display HLA distribution patterns different and sometimes opposite from areas of low incidence, whereas populations in areas with intermediate incidence display a totally independent pattern. Two main reasons may explain this association between HLA phenotype distribution and the risk of developing NPC in various populations. First, given the fact that expression of Epstein-Barr Virus (EBV) proteins by cancer cells is tightly linked with NPC development, HLA may influence the development of NPC by modulating the expression of EBV proteins. This explanation is, however, based primarily on theoretical assumptions given that no clear definition of HLA binding pattern of EBV epitopes has been directly shown to significantly alter the recognition of EBV proteins and the risk of developing the disease. Alternatively, HLA may represent a genetic marker flagging the presence of a NPC predisposition locus in close linkage disequilibrium with the HLA class I region. A critical review of known HLA associations in various geographical areas and their interpretation will be presented in this review.
Collapse
Affiliation(s)
- Xin Li
- Infectious Disease and Immunogenetics Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | |
Collapse
|
15
|
Merlo A, Turrini R, Dolcetti R, Zanovello P, Amadori A, Rosato A. Adoptive cell therapy against EBV-related malignancies: a survey of clinical results. Expert Opin Biol Ther 2008; 8:1265-94. [PMID: 18694349 DOI: 10.1517/14712598.8.9.1265] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Epstein-Barr Virus (EBV) infection is associated with a heterogeneous group of tumors, including lymphoproliferative disorders, Hodgkin's disease, nasopharyngeal carcinoma and Burkitt's lymphoma. As such neoplastic disorders express viral antigens, they can be treated by adoptive immunotherapy strategies relying mostly on in vitro generation and expansion of virus-specific cytotoxic T lymphocytes (CTL), which can be administered to patients for both prophylaxis and treatment. OBJECTIVE We reviewed results obtained in all clinical trials reported thus far employing anti-EBV adoptive immunotherapy for different virus-related malignancies. METHODS 'PTLD after HSCT', 'PTLD after SOT', 'NPC', 'HD', 'SCAEBV' and 'extranodal NK/T cell lymphoma', in combination with 'Adoptive immunotherapy' and 'Adoptive transfer', were used as search keys for papers in PubMed. CONCLUSIONS Although the heterogeneity of different studies precludes their collection for a meta-analysis, it can be inferred that adoptive therapy with EBV-specific CTL is safe, well tolerated and particularly effective in the case of most immunogenic tumors, like post-transplant lymphoproliferative disease.
Collapse
Affiliation(s)
- Anna Merlo
- University of Padova, Department of Oncology and Surgical Sciences, Via Gattamelata 64, I-35128 Padova, Italy
| | | | | | | | | | | |
Collapse
|
16
|
Davis JE, Moss DJ. Epstein-Barr virus vaccines. Vaccines (Basel) 2008. [DOI: 10.1016/b978-1-4160-3611-1.50050-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
17
|
Abstract
Clinical trials have established that T cells have the ability to prevent and treat pathogens and tumors. This is perhaps best exemplified by engraftment of allogeneic T cells in the context of hematopoietic stem-cell transplantation (HSCT), which for over the last 50 years remains one of the best and most robust examples of cell-based therapies for the treatment of hematologic malignancies. Yet, the approach to infuse T cells for treatment of cancer, in general, and pediatric tumors, in particular, generally remains on the sidelines of cancer therapy. This review outlines the current state-of-the-art and provides a rationale for undertaking adoptive immunotherapy trials with emphasis on childhood malignancies.
Collapse
|
18
|
Liu FF, Frappier L, Kim J, O'Sullivan B, Hui A, Bastianutto C. East-West Symposium on nasopharyngeal cancer. Int J Radiat Oncol Biol Phys 2007; 67:703-8. [PMID: 17141974 DOI: 10.1016/j.ijrobp.2006.09.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2006] [Accepted: 09/06/2006] [Indexed: 10/23/2022]
Abstract
BACKGROUND To achieve greater understanding of the epidemiology, pathogenesis, molecular oncology, diagnostic, and therapeutic aspects of nasopharyngeal cancer (NPC), an international meeting was held in June 2005, Toronto, Canada. RESULTS Further insights were obtained into the role of EBV in NPC development, with its diverse effects ranging from proliferative signals via NF-kB, to immunesuppression, to angiogenic gene regulation. Subsequently, multiple pathways are dysregulated in NPC as revealed by expression array analyses, including apoptosis, integrin, and B-catenin cascades. Advances have been made in the diagnosis and monitoring of NPC, using transoral brushings and plasma levels of EBV transcripts, which may not directly correlate with the number of circulating tumor cells, but is nevertheless informative in predicting and tracking disease response. Many novel therapies have promising results, particularly in the areas of immunotherapies, and the exploration of molecularly targeted approaches such as cetuximab or histone deacetylase inhibitors. CONCLUSIONS The results from large randomized trials and meta-analyses have consistently demonstrated the benefit of concurrent chemotherapy with curative radiation therapy, but at a cost of greater acute and late-tissue toxicities. Further advances are required to achieve an improved understanding on the inter-relationship between environmental and genetic determinants in NPC development, to reduce the global burden of this disease. At the same time, novel therapeutic approaches are necessary to increase curability of NPC, but with reduced long-term toxicities.
Collapse
Affiliation(s)
- Fei-Fei Liu
- Department of Radiation Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada.
| | | | | | | | | | | |
Collapse
|