1
|
Martinov T, Greenberg PD. Targeting Driver Oncogenes and Other Public Neoantigens Using T Cell Receptor-Based Cellular Therapy. ANNUAL REVIEW OF CANCER BIOLOGY 2023; 7:331-351. [PMID: 37655310 PMCID: PMC10470615 DOI: 10.1146/annurev-cancerbio-061521-082114] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
T cell reactivity to tumor-specific neoantigens can drive endogenous and therapeutically induced antitumor immunity. However, most tumor-specific neoantigens are unique to each patient (private) and targeting them requires personalized therapy. A smaller subset of neoantigens includes epitopes that span recurrent mutation hotspots, translocations, or gene fusions in oncogenic drivers and tumor suppressors, as well as epitopes that arise from viral oncogenic proteins. Such antigens are likely to be shared across patients (public), uniformly expressed within a tumor, and required for cancer cell survival and fitness. Although a limited number of these public neoantigens are naturally immunogenic, recent studies affirm their clinical utility. In this review, we highlight efforts to target mutant KRAS, mutant p53, and epitopes derived from oncogenic viruses using T cells engineered with off-the-shelf T cell receptors. We also discuss the challenges and strategies to achieving more effective T cell therapies, particularly in the context of solid tumors.
Collapse
Affiliation(s)
- Tijana Martinov
- Program in Immunology and Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Philip D Greenberg
- Program in Immunology and Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Immunology Department, University of Washington, Seattle, Washington, USA
| |
Collapse
|
2
|
Meissner J, Schmitt M, Andrulis M, Schweizer L, Dietrich S, Alber B, Harting I, Kurz FT, Martens UM, Ho AD, Müller-Tidow C, Dreger P. Cure of intravascular NK/T-cell lymphoma of the central nervous system by allogeneic hematopoietic cell transplantation. Bone Marrow Transplant 2022; 57:1451-1454. [PMID: 35680996 PMCID: PMC9439947 DOI: 10.1038/s41409-022-01734-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Julia Meissner
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany.
| | - Michael Schmitt
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Mindaugas Andrulis
- Department of General Pathology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Institute of Pathology, Klinikum Ludwigshafen, Ludwigshafen, Germany
| | - Leonille Schweizer
- Department of Neuropathology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Institute of Neuropathology, Charité, Berlin, Germany
| | - Sascha Dietrich
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Bettina Alber
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Inga Harting
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany
| | - Felix T Kurz
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany.,Department of Radiology, German Cancer Research Center, Heidelberg, Germany
| | - Uwe M Martens
- Department of Hematology/Oncology, Clinics Heilbronn GmbH, Heilbronn, Germany
| | - Anthony D Ho
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | - Peter Dreger
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
3
|
Zhang C, Tan Q, Li S, Shen L, Zhang J, Liu Y, Yang W, Lu Z. Induction of EBV latent membrane protein-2A (LMP2A)-specific T cells and construction of individualized TCR-engineered T cells for EBV-associated malignancies. J Immunother Cancer 2021; 9:jitc-2021-002516. [PMID: 34210819 PMCID: PMC8252876 DOI: 10.1136/jitc-2021-002516] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2021] [Indexed: 12/05/2022] Open
Abstract
Background Latent membrane protein-2A (LMP2A)-specific TCR-engineered T cells could be a promising treatment approach to Epstein–Barr virus-associated malignancies. However, previous studies mainly reported LMP2A-reactive TCRs only focusing on specific HLA subtypes and corresponding epitopes, and thus, they were only suitable for patients with specific HLA. Methods Due to hugely varied HLA subtypes and presented LMP2A epitopes in different individuals, our study attempted to develop an individualized approach, based on the weekly in vitro stimulation of peripheral T cells for 2 weeks with autologous dendritic cells (DCs) pulsed with a pool of LMP2A peptides covering LMP2A whole protein and combination analysis of high throughput TCRβ sequencing of prestimulated and poststimulated T cells and single-cell TCR sequencing of poststimulated T cells, and to identify LMP2A-specific TCRs of which poststimulated frequencies significantly increased than corresponding prestimulated frequencies. Results Following this approach, multiple LMP2A-reactive TCRs were identified, optimized and cloned into lentiviral vector, and then transduced into peripheral T cells. These engineerd T cells were demonstrated to specifically recognize the LMP2A presented by autologous DCs and lymphoblastoid cell lines in vitro and in vivo. Conclusions This approach provides an efficient procedure to isolate individualized LMP2A-specific TCRs for basic and translational research, as well as for clinical applications.
Collapse
Affiliation(s)
- Chaoting Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qin Tan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shance Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Luyan Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jingtao Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ying Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wenjun Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China .,Key Laboratory of Fertility Preservation and Maintenance, School of Basic Medicine and the General Hospital, Ningxia Medical University, Yinchuan, China
| | - Zheming Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
4
|
Abstract
An important role of the immune system is in the surveillance for abnormal or transformed cells, which is known as cancer immunosurveillance. Through this process, the first changes to normal tissue homeostasis caused by infectious or other inflammatory insults can be detected by the immune system through the recognition of antigenic molecules (including tumour antigens) expressed by abnormal cells. However, as they develop, tumour cells can acquire antigenic and other changes that allow them to escape elimination by the immune system. To bias this process towards elimination, immunosurveillance can be improved by the administration of vaccines based on tumour antigens. Therapeutic cancer vaccines have been extensively tested in patients with advanced cancer but have had little clinical success, which has been attributed to the immunosuppressive tumour microenvironment. Thus, the administration of preventive vaccines at pre-malignant stages of the disease holds promise, as they function before tumour-associated immune suppression is established. Accordingly, immunological and clinical studies are yielding impressive results.
Collapse
|
5
|
Th17 immune microenvironment in Epstein-Barr virus-negative Hodgkin lymphoma: implications for immunotherapy. Blood Adv 2017; 1:1324-1334. [PMID: 29296775 DOI: 10.1182/bloodadvances.2017007260] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/21/2017] [Indexed: 12/15/2022] Open
Abstract
Classical Hodgkin lymphoma (CHL) is a neoplasm characterized by robust inflammatory infiltrates and heightened expression of the immunosuppressive PD-1/PD-L1 pathway. Although anti-PD-1 therapy can be effective in >60% of patients with refractory CHL, improved treatment options are needed for CHLs which are resistant to anti-PD-1 or relapse after this form of immunotherapy. A deeper understanding of immunologic factors in the CHL microenvironment might support the design of more effective treatment combinations based on anti-PD-1. In addition, because the Epstein-Barr virus (EBV) residing in some CHL tumors is strongly immunogenic, we hypothesized that characteristics of the tumor immune microenvironment in EBV+ CHL would be distinct from EBV- CHL, with specific implications for designing combination treatment regimens. Employing immunohistochemistry for immune cell subsets and checkpoint molecules, as well as gene expression profiling, we characterized 32 CHLs from the Johns Hopkins archives, including 12 EBV+ and 20 EBV- tumors. Our results revealed a dichotomous cellular and cytokine immune milieu in EBV+ vs EBV- CHL. EBV+ tumors displayed a T helper 1 (Th1) profile typical of effective antitumor immunity, with increased infiltration of CD8+ T cells and coordinate expression of the canonical Th1 transcription factor Tbet (TBX21), interferon-γ (IFNG), and the IFN-γ-inducible immunosuppressive enzyme indoleamine 2,3-dioxygenase. In contrast, EBV- tumors manifested a pathogenic Th17 profile and ongoing engagement of the interleukin-23 (IL-23)/IL-17 axis, with heightened phosphorylated signal transducer and activator of transcription 3 expression in infiltrating lymphocytes. These findings suggest that drugs blocking the IL-23/IL-17 axis, which are already in the clinic for treating certain autoimmune disorders, may enhance the therapeutic impact of anti-PD-1 therapy in EBV- CHL.
Collapse
|
6
|
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: 3.7] [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
|
7
|
McLaughlin LP, Gottschalk S, Rooney CM, Bollard CM. EBV-Directed T Cell Therapeutics for EBV-Associated Lymphomas. Methods Mol Biol 2017; 1532:255-265. [PMID: 27873282 DOI: 10.1007/978-1-4939-6655-4_19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Epstein Barr virus (EBV) is a human gamma herpes virus that establishes latency in B cells after primary infection. EBV generally only causes a mild, self-limiting viral illness but is also associated with several malignancies including posttransplantation lymphoproliferative disorder in the immunosuppressed host as well as Hodgkin and non-Hodgkin lymphoma in the immune competent host. The expression of EBV antigens by lymphoma has important applications as targets for adoptive T cell therapy. However, as many lymphomas only express subdominant EBV antigens that are less immunogenic, novel strategies are needed to manufacture EBV-specific T cell products specific for Latent Membrane Protein 1 (LMP1) and LMP2, which are expressed in lymphomas with type II and III latency. While several techniques for manufacturing EBV-CTLs are described in the literature, this chapter focuses on one method for generating Good Manufacturing Practice (GMP)-compliant EBV-specific T cell products that are enriched with LMP1 and LMP2.
Collapse
Affiliation(s)
- Lauren P McLaughlin
- Department of Hematology/Oncology, Children's National Medical Center, 111 Michigan Ave., Washington, DC, 20010, USA.
- The George Washington University, Washington, DC, USA.
| | - Stephen Gottschalk
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital,, Baylor College of Medicine, Houston, TX, USA
| | - Catherine M Bollard
- The George Washington University, Washington, DC, USA
- Center for Cancer and Immunology Research, Children's National Medical Center, Washington, DC, USA
| |
Collapse
|
8
|
Barth MJ, Chu Y, Hanley PJ, Cairo MS. Immunotherapeutic approaches for the treatment of childhood, adolescent and young adult non-Hodgkin lymphoma. Br J Haematol 2016; 173:597-616. [PMID: 27062282 DOI: 10.1111/bjh.14078] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
With the introduction of the anti-CD20 monoclonal antibody rituximab, B-cell non-Hodgkin lymphoma was the first malignancy successfully treated with an immunotherapeutic agent. Since then, numerous advances have expanded the repertoire of immunotherapeutic agents available for the treatment of a variety of malignancies, including many lymphoma subtypes. These include the introduction of monoclonal antibodies targeting a variety of cell surface proteins, including the successful targeting of immunoregulatory checkpoint receptors present on T-cells or tumour cells. Additionally, cellular immunotherapeutic approaches utilize T- or Natural Killer-cells generated with chimeric antigen receptors against cell surface proteins or Epstein-Barr virus-associated latent membrane proteins. The following review describes the current state of immunotherapy for non-Hodgkin lymphoma including a summary of currently available data and promising agents currently in clinical development with future promise in the treatment of childhood, adolescent and young adult non-Hodgkin lymphoma.
Collapse
Affiliation(s)
- Matthew J Barth
- Department of Pediatrics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Division of Pediatric Hematology/Oncology, University at Buffalo, Buffalo, NY, USA
| | - Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - Patrick J Hanley
- Program for Cell Enhancement and Technologies for Immunotherapy, Division of Blood and Marrow Transplantation, Sheikhz Zayed Institute for Pediatric Surgical Innovation, Washington, D.C., USA.,Center for Cancer and Immunology Research, Children's National Health System, The George Washington University, Washington, D.C., USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA.,Department of Medicine, New York Medical College, Valhalla, NY, USA.,Department of Pathology, New York Medical College, Valhalla, NY, USA.,Department of Microbiology & Immunology, New York Medical College, Valhalla, NY, USA.,Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
9
|
Deng R, Yi H, Liu YL, Fan FY, Fu LI, Li YC, Li GS, Lai SH, Miao XJ, Shuai YR, He GC, Wang YI, Zeng Y, Sun HP, Qiu L, Su YI. Enhanced antitumor effect of combining chemotherapy with Epstein-Barr virus (EBV)-specific cytotoxic T lymphocytes in mice with EBV-related non-Hodgkin's lymphoma. Mol Clin Oncol 2016; 3:1233-1238. [PMID: 26807226 DOI: 10.3892/mco.2015.646] [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: 06/02/2015] [Accepted: 08/28/2015] [Indexed: 11/05/2022] Open
Abstract
Epstein-Barr virus (EBV)-related non-Hodgkin's lymphoma (NHL) represents a major problem in hematological clinical studies due to its drug tolerance and refractoriness. EBV infection is a key factor driving the process of tumor growth. Immune therapy is an important biotherapeutic method of treating cancer, which is attracting increasing attention. We hypothesized that combining conventional chemotherapy with immune therapy in the treatment of EBV-related NHL may achieve better outcomes. First, we successfully cloned large numbers of EBV-specific T cells by immune stimulation ex vivo. Subsequently, the combined therapy was applied in a murine model of human EBV-related NHL. As expected, combined therapy inhibited tumor growth more effectively compared with monotherapy. In addition, we continuously tested the tumor-associated immune microenvironment and observed that the numbers of tumor-infiltrating cytotoxic T lymphocytes (CTLs) and macrophages were elevated following combined therapy. These effects suggest that EBV-specific CTLs may indirectly promote an innate immune reaction in lymphoma by activating tumor-infiltrating macrophage proliferation. Our findings may provide a guide for the prospective treatment of EBV-related NHL.
Collapse
Affiliation(s)
- Rui Deng
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Hai Yi
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Yi-Lan Liu
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Fang-Yi Fan
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - L I Fu
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Ye-Cheng Li
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Guo-Shun Li
- Department of Translational Medicine, Experimental Medical Research Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Si-Han Lai
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Xiao-Juan Miao
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Yan-Rong Shuai
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Guang-Cui He
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Y I Wang
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Yan Zeng
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Hao-Ping Sun
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Ling Qiu
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Y I Su
- Department of Hematology, Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Cheng Du Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| |
Collapse
|
10
|
Epstein Barr virus-associated lymphoproliferative diseases: the virus as a therapeutic target. Exp Mol Med 2015; 47:e136. [PMID: 25613733 PMCID: PMC4314579 DOI: 10.1038/emm.2014.102] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 10/15/2014] [Indexed: 02/07/2023] Open
Abstract
Epstein Barr virus (EBV)-associated lymphoproliferative diseases (LPDs) express all EBV latent antigens (type III latency) in immunodeficient patients and limited antigens (type I and II latencies) in immunocompetent patients. Post-transplantation lymphoproliferative disease (PTLD) is the prototype exhibiting type III EBV latency. Although EBV antigens are highly immunogenic, PTLD cell proliferation remains unchecked because of the underlying immunosuppression. The restoration of anti-EBV immunity by EBV-specific T cells of either autologous or allogeneic origin has been shown to be safe and effective in PTLDs. Cellular therapy can be improved by establishing a bank of human leukocyte antigen-characterized allogeneic EBV-specific T cells. In EBV+ LPDs exhibiting type I and II latencies, the use of EBV-specific T cells is more limited, although the safety and efficacy of this therapy have also been demonstrated. The therapeutic role of EBV-specific T cells in EBV+ LPDs needs to be critically reappraised with the advent of monoclonal antibodies and other targeted therapy. Another strategy involves the use of epigenetic approaches to induce EBV to undergo lytic proliferation when expression of the viral thymidine kinase renders host tumor cells susceptible to the cytotoxic effects of ganciclovir. Finally, the prophylactic use of antiviral drugs to prevent EBV reactivation may decrease the occurrence of EBV+ LPDs.
Collapse
|
11
|
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
|
12
|
Emery VC, Einsele H, Atabani S, Haque T. Immunotherapy and vaccination after transplant: the present, the future. Hematol Oncol Clin North Am 2011; 25:215-29. [PMID: 21236399 DOI: 10.1016/j.hoc.2010.11.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Vaccination and adoptive immunotherapy for herpes virus infections has become an attractive option for the control of a virus family that negatively affects transplantation. In the future, enhanced ability to select antigen-specific T cells without significant in vitro manipulation should provide new opportunities for refining and enhancing adoptive immunotherapeutic approaches. This article focuses on advances in the area of vaccinology for some of these infections and in the use of adoptive immunotherapy. At present, many of these approaches in transplant recipients have focused on infections such as human cytomegalovirus, but the opportunity to use these examples as proof of concept for other infections is discussed.
Collapse
Affiliation(s)
- Vincent C Emery
- Department of Infection (Royal Free Campus), University College London, Rowland Hill Street, Hampstead, London, UK
| | | | | | | |
Collapse
|
13
|
Emery VC, Einsele H, Atabani S, Haque T. Immunotherapy and vaccination after transplant: the present, the future. Infect Dis Clin North Am 2010; 24:515-29. [PMID: 20466281 DOI: 10.1016/j.idc.2010.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Vaccination and adoptive immunotherapy for herpes virus infections has become an attractive option for the control of a virus family that negatively affects transplantation. In the future, enhanced ability to select antigen-specific T cells without significant in vitro manipulation should provide new opportunities for refining and enhancing adoptive immunotherapeutic approaches. This article focuses on advances in the area of vaccinology for some of these infections and in the use of adoptive immunotherapy. At present, many of these approaches in transplant recipients have focused on infections such as human cytomegalovirus, but the opportunity to use these examples as proof of concept for other infections is discussed.
Collapse
Affiliation(s)
- Vincent C Emery
- Department of Infection, University College London, Rowland Hill Street, Hampstead, London NW3 2QG, UK.
| | | | | | | |
Collapse
|
14
|
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
|
15
|
Diekmann J, Adamopoulou E, Beck O, Rauser G, Lurati S, Tenzer S, Einsele H, Rammensee HG, Schild H, Topp MS. Processing of two latent membrane protein 1 MHC class I epitopes requires tripeptidyl peptidase II involvement. THE JOURNAL OF IMMUNOLOGY 2009; 183:1587-97. [PMID: 19587004 DOI: 10.4049/jimmunol.0803441] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The EBV Ag latent membrane protein 1 (LMP1) has been described as a potential target for T cell immunotherapy in EBV-related malignancies. However, only a few CD8(+) T cell epitopes are known, and the benefit of LMP1-specific T cell immunotherapy has not yet been proven. In this work, we studied the processing of the two LMP1 HLA-A02-restricted epitopes, YLLEMLRWL and YLQQNWWTL. We found that target cells endogenously expressing the native LMP1 are not recognized by CTLs specific for these epitopes because the N-terminal part of LMP1 limits the efficiency of epitope generation. We further observed that the proteasome is not required for the generation of both epitopes and that the YLLEMLRWL epitope seems to be destroyed by the proteasome, because blocking of proteasomal activities enhanced specific CTL activation. Activation of LMP1-specific CTLs could be significantly reduced after inhibition of the tripeptidyl peptidase II, suggesting a role for this peptidase in the processing of both epitopes. Taken together, our results demonstrate that the MHC class I-restricted LMP1 epitopes studied in this work are two of very few epitopes known to date to be processed proteasome independently by tripeptidyl peptidase II.
Collapse
Affiliation(s)
- Jan Diekmann
- Medical Clinic und Policlinic II, Julius-Maximilian University of Wuerzburg, Wuerzburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kasamon YL, Ambinder RF. Immunotherapies for Hodgkin's lymphoma. Crit Rev Oncol Hematol 2008; 66:135-44. [PMID: 18023356 PMCID: PMC5792053 DOI: 10.1016/j.critrevonc.2007.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2007] [Revised: 09/19/2007] [Accepted: 10/03/2007] [Indexed: 11/17/2022] Open
Abstract
Multiple immune evasion strategies characterize the pathobiology of Hodgkin's lymphoma. These must be considered when developing and testing immunotherapeutic approaches for this disease. The clinical experience with adoptive immunotherapy of Epstein-Barr virus positive tumors, and with monoclonal antibodies directed against CD30, CD20, and other antigens, is herein reviewed.
Collapse
Affiliation(s)
- Yvette L Kasamon
- Division of Hematologic Malignancies, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | | |
Collapse
|
17
|
Moskowitz CH. An Evidence-Based Approach to the Management of Hodgkin’s Lymphoma. Oncology 2007. [DOI: 10.1007/0-387-31056-8_67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
18
|
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
|
19
|
In vitro methods for generating highly purified EBV associated tumor antigen-specific T cells by using solid phase T cell selection system for immunotherapy. J Immunol Methods 2007; 328:169-81. [PMID: 17931649 DOI: 10.1016/j.jim.2007.08.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2007] [Revised: 08/06/2007] [Accepted: 08/29/2007] [Indexed: 12/13/2022]
Abstract
Adoptive cell transfer immunotherapy has been utilized to treat EBV related human malignancies including post-transplant lymphoproliferative diseases, Hodgkin's lymphoma and nasopharyngeal carcinoma. However, there are limited options available for tumor antigen-specific T cell purification. Here we describe a novel solid phase T cell selection system, in which monocytes or EBV transformed B-lymphocytes are immobilized on solid support for antigen-specific T cell purification. We hypothesize and prove that antigen-specific T cells recognize their cognate antigens and bind to them faster than non-antigen specific T cells. Therefore antigen-specific T cells can be concentrated on the surface after removing the non-adherent cells by washing. The optimal selection time for both EBV-specific T cells and LMP2-specific T cells is studied. Our data demonstrate that the frequency of antigen-specific T cells can be increased by >20-fold after selection. Moreover, activated antigen-specific T cells proliferate more rapidly than non-specific T cells, further increasing the frequency and purity of antigen-specific T cells. This new T cell selection system is superior to traditional repeated stimulation methods in generating tumor antigen-specific T cells. We are able to generate large quantities of highly purified T cells of subdominant antigens LMP2 within 2 weeks after T cell activation for adoptive cell transfer immunotherapy with this simple, rapid and inexpensive T cell selection system.
Collapse
|
20
|
Sun Q, Brewer N, Dunham K, Chen L, Bao L, Burton R, Lucas KG. Interferon-gamma expressing EBV LMP2A-specific T cells for cellular immunotherapy. Cell Immunol 2007; 246:81-91. [PMID: 17663985 DOI: 10.1016/j.cellimm.2007.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2007] [Revised: 06/12/2007] [Accepted: 06/13/2007] [Indexed: 11/29/2022]
Abstract
To generate therapeutic T cells for adoptive immunotherapy, T cells specific to Epstein-Barr virus LMP2A were enriched on the basis of antigen-specific production of interferon-gamma (IFNgamma). The enriched T cells, contained over 60% LMP2A-specific effectors, were polyclonal and targeted multiple LMP2A epitopes. A high proportion of the enriched T cells produced the Th1 cytokines interleukin (IL)-2 and granulocyte monocyte colony stimulating factor, while few cells expressed the Th2 cytokines IL4 and IL10. The enriched T cells specifically lysed LMP2A-expressing target cells, with concomitant production of IFNgamma and surface expression of CD107, suggesting the involvement of the granule exocytosis-mediated cytolytic pathway. In addition, the enriched T cells expressed CD45RO, CD28 and CD27, but not CD45RA, consistent with a differentiation stage capable of self-renewal for long-term persistence. LMP2A-specific T cells enriched based on IFNgamma-production may provide improved efficacy for the treatment of Epstein-Barr virus related malignancy.
Collapse
Affiliation(s)
- Qi Sun
- Division of Hematology/Oncology, Department of Pediatrics, Pennsylvania State College of Medicine, Pennsylvania State Milton S. Hershey Medical Center, Pennsylvania State Children's Hospital, 500 University Drive, Hershey, PA 17033-0850, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Adoptive transfer of tumor-specific T-cells is an attractive strategy for the treatment of patients with refractory or relapsed Hodgkin's lymphoma. However, Hodgkin's lymphomas possess a range of tumor-evasion mechanisms, which must be overcome before the full potential of immunotherapies can be achieved. In this article, we discuss the promise of Epstein-Barr virus-specific cytotoxic T-lymphocytes, the roles of cytokines, and other strategies for overcoming the immune-evasion mechanisms in Hodgkin's lymphoma.
Collapse
Affiliation(s)
- Alana A Kennedy-Nasser
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, Texas 77030, USA
| | | | | |
Collapse
|
22
|
Cho HI, Hong YS, Lee MA, Kim EK, Yoon SH, Kim CC, Kim TG. Adoptive transfer of Epstein-Barr virus-specific cytotoxic T-lymphocytes for the treatment of angiocentric lymphomas. Int J Hematol 2006; 83:66-73. [PMID: 16443556 DOI: 10.1532/ijh97.a30505] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Angiocentric lymphoma, known as natural killer (NK)/T-cell non-Hodgkin's lymphoma, has been reported to be associated with the Epstein-Barr virus (EBV). We performed adoptive transfer of EBV-specific polyclonal T-cell lines in 3 patients with extranodal NK/T-cell lymphoma, nasal type, and evaluated the treatment for safety, immunologic reconstitution, and clinical outcomes. The tissue samples collected from the 3 patients were confirmed by polymerase chain reaction analysis to be EBV positive. In the cases of the first and second patients, EBV-transformed B-lymphoblastoid cell lines (LCLs) and T-cell lines were generated from peripheral lymphocytes of HLA-matched sibling donors. The third patient's T-cell lines were induced with autologous lymphocytes. Polyclonal T-cell infusion was carried out after high-dose radiotherapy because active relapsed disease remained in all of the patients. The first patient received 4 weekly infusions of 2 3 10(7) cells/m(2), and the second and third patients underwent treatment with 2 cycles of infusions of the same dosage. All T-cell lines showed >60% NK activity, cytotoxic T-lymphocyte (CTL) responses of >40% against autologous LCLs, and no CTL activity against patient-derived lymphoblasts. The level of cytotoxicity increased substantially in all patients after cell infusion. The 2 patients who received T-cell therapy twice had stabilized disease for more than 3 years. These safe treatments exhibited no severe inflammatory response, and no serious toxicity developed during T-cell therapy. Our findings demonstrate that adoptively transferred cells may provide reconstitution of EBV-specific CTL responses in patients with active relapsed angiocentric lymphoma. These results provide a rationale for the immunotherapy of angiocentric lymphoma.
Collapse
MESH Headings
- Adult
- Cell Line
- Disease-Free Survival
- Epstein-Barr Virus Infections/pathology
- Epstein-Barr Virus Infections/therapy
- Herpesvirus 4, Human
- Humans
- Immunotherapy, Adoptive/methods
- Killer Cells, Natural/pathology
- Killer Cells, Natural/virology
- Lymphoma, Non-Hodgkin/pathology
- Lymphoma, Non-Hodgkin/therapy
- Lymphoma, Non-Hodgkin/virology
- Male
- Nose Neoplasms/pathology
- Nose Neoplasms/therapy
- Nose Neoplasms/virology
- T-Lymphocytes, Cytotoxic/pathology
- T-Lymphocytes, Cytotoxic/transplantation
- T-Lymphocytes, Cytotoxic/virology
- Treatment Outcome
Collapse
Affiliation(s)
- Hyun-Il Cho
- Department of Microbiology and Immunology, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seocho-Ku, Seoul, 137-701, Korea
| | | | | | | | | | | | | |
Collapse
|
23
|
Bollard CM, Huls MH, Buza E, Weiss H, Torrano V, Gresik MV, Chang J, Gee A, Gottschalk SM, Carrum G, Brenner MK, Rooney CM, Heslop HE. Administration of latent membrane protein 2-specific cytotoxic T lymphocytes to patients with relapsed Epstein-Barr virus-positive lymphoma. ACTA ACUST UNITED AC 2006; 6:342-7. [PMID: 16507214 DOI: 10.3816/clm.2006.n.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Catherine M Bollard
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Comoli P, Rooney C. Treatment of Epstein–Barr Virus Infections: Chemotherapy, Antiviral Therapy, and Immunotherapy. EPSTEIN-BARR VIRUS 2006. [DOI: 10.3109/9781420014280.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
25
|
Straathof KC, Leen AM, Buza EL, Taylor G, Huls MH, Heslop HE, Rooney CM, Bollard CM. Characterization of latent membrane protein 2 specificity in CTL lines from patients with EBV-positive nasopharyngeal carcinoma and lymphoma. THE JOURNAL OF IMMUNOLOGY 2005; 175:4137-47. [PMID: 16148164 DOI: 10.4049/jimmunol.175.6.4137] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Viral proteins expressed by EBV-associated tumors provide target Ags for immunotherapy. Adoptive T cell therapy has proven effective for posttransplant EBV-associated lymphoma in which all EBV latent Ags are expressed (type III latency). Application of immunotherapeutic strategies to tumors such as nasopharyngeal carcinoma and Hodgkin's lymphoma that have a restricted pattern of EBV Ag expression (type II latency) is under investigation. Potential EBV Ag targets for T cell therapy expressed by these tumors include latent membrane proteins (LMP) 1 and 2. A broad panel of epitopes must be identified from these target Ags to optimize vaccination strategies and facilitate monitoring of tumor-specific T cell populations after immunotherapeutic interventions. To date, LMP2 epitopes have been identified for only a limited number of HLA alleles. Using a peptide library spanning the entire LMP2 sequence, 25 CTL lines from patients with EBV-positive malignancies expressing type II latency were screened for the presence of LMP2-specific T cell populations. In 21 of 25 lines, T cell responses against one to five LMP2 epitopes were identified. These included responses to previously described epitopes as well as to newly identified HLA-A*0206-, A*0204/17-, A29-, A68-, B*1402-, B27-, B*3501-, B53-, and HLA-DR-restricted epitopes. Seven of the nine newly identified epitopes were antigenically conserved among virus isolates from nasopharyngeal carcinoma tumors. These new LMP2 epitopes broaden the diversity of HLA alleles with available epitopes, and, in particular, those epitopes conserved between EBV strains provide valuable tools for immunotherapy and immune monitoring.
Collapse
Affiliation(s)
- Karin C Straathof
- Center for Cell and Gene Therapy, Baylor College of Medicine, Methodist Hospital and Texas Children's Hospital, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Majority of patients with Hodgkin lymphoma are cured with current therapy. The short and long-term toxicity of therapy is a current issue, most especially the radiation-induced second tumors and cardiac abnormalities. Optimal therapy of advanced disease is being compared to ABVD (doxorubicin, bleomycin, vinblastine, dacarbazine). More intensive chemotherapy may result in higher response but greater toxicity. Regimens requiring radiotherapy are also at risk of long-term toxicity. New targeted biologic therapy is under current investigation.
Collapse
Affiliation(s)
- George P Canellos
- Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Claviez A, Tiemann M, Lüders H, Krams M, Parwaresch R, Schellong G, Dörffel W. Impact of latent Epstein-Barr virus infection on outcome in children and adolescents with Hodgkin's lymphoma. J Clin Oncol 2005; 23:4048-56. [PMID: 15961758 DOI: 10.1200/jco.2005.01.701] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The prognostic significance of latent Epstein-Barr virus (EBV) infection in Hodgkin's lymphoma (HL) is debated controversially. Especially in the pediatric age group, no conclusive data are available. PATIENTS AND METHODS Eight hundred forty-two children and adolescents (median age, 13.7 years) from pediatric multicenter treatment studies HD-90 and HD-95 were studied for latent EBV infection in Hodgkin's and Reed-Sternberg cells by immunostaining against latent membrane protein 1 (LMP-1). Results were compared with established risk factors. RESULTS Two hundred sixty-three patients (31%) were LMP positive. EBV infection correlated with sex (39% male v 23% female; P < .001), histologic subtype (69% mixed cellularity v 22% nodular sclerosis v 6% lymphocyte predominance; P < .001) and young age. With a median follow-up of 4.9 years, 820 patients (97%) are alive. Probability of overall survival at 10 years (+/- standard deviation) for EBV-negative and -positive patients was 98.1% +/- 0.6% and 95.1% +/- 1.4%, respectively (P = .017 by log-rank test). A negative effect of EBV infection became evident for patients with nodular sclerosis subtype Bennett II (P = .02), and those treated for advanced stages (P = .003). In multivariate analysis, LMP positivity was an independent factor for adverse outcome (RR = 3.08). Probability of failure-free survival (FFS) in LMP positive and negative patients was 89.1% +/- 2.3% and 84.1% +/- 3.9%, respectively (P = .86). CONCLUSION With effective combined treatment modalities in pediatric HL, latent EBV infection has no influence on FFS but is associated with an inferior overall survival in crucial subgroups.
Collapse
Affiliation(s)
- Alexander Claviez
- Department of Pediatrics, Germany Lymph Registry, University of Kiel, Schwanenweg 20, 24105 Kiel, Germany.
| | | | | | | | | | | | | |
Collapse
|
28
|
Bollard CM, Aguilar L, Straathof KC, Gahn B, Huls MH, Rousseau A, Sixbey J, Gresik MV, Carrum G, Hudson M, Dilloo D, Gee A, Brenner MK, Rooney CM, Heslop HE. Cytotoxic T lymphocyte therapy for Epstein-Barr virus+ Hodgkin's disease. ACTA ACUST UNITED AC 2005; 200:1623-33. [PMID: 15611290 PMCID: PMC2211993 DOI: 10.1084/jem.20040890] [Citation(s) in RCA: 285] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Epstein Barr virus (EBV)+ Hodgkin's disease (HD) expresses clearly identified tumor antigens derived from the virus and could, in principle, be a target for adoptive immunotherapy with viral antigen–specific T cells. However, like most tumor-associated antigens in immunocompetent hosts, these potential targets are only weakly immunogenic, consisting primarily of the latent membrane protein (LMP)1 and LMP2 antigens. Moreover, Hodgkin tumors possess a range of tumor evasion strategies. Therefore, the likely value of immunotherapy with EBV-specific cytotoxic effector cells has been questioned. We have now used a combination of gene marking, tetramer, and functional analyses to track the fate and assess the activity of EBV cytotoxic T lymphocyte (CTL) lines administered to 14 patients treated for relapsed EBV+ HD. Gene marking studies showed that infused effector cells could further expand by several logs in vivo, contribute to the memory pool (persisting up to 12 mo), and traffic to tumor sites. Tetramer and functional analyses showed that T cells reactive with the tumor-associated antigen LMP2 were present in the infused lines, expanded in peripheral blood after infusion, and also entered tumor. Viral load decreased, demonstrating the biologic activity of the infused CTLs. Clinically, EBV CTLs were well tolerated, could control type B symptoms (fever, night sweats, and weight loss), and had antitumor activity. After CTL infusion, five patients were in complete remission at up to 40 mo, two of whom had clearly measurable tumor at the time of treatment. One additional patient had a partial response, and five had stable disease. The performance and fate of these human tumor antigen–specific T cells in vivo suggests that they might be of value for the treatment of EBV+ Hodgkin lymphoma.
Collapse
Affiliation(s)
- Catherine M Bollard
- Center for Gene and Cell Therapy, Baylor College of Medicine, 6621 Fannin St., MC 3-3320, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Bollard CM, Straathof KCM, Huls MH, Leen A, Lacuesta K, Davis A, Gottschalk S, Brenner MK, Heslop HE, Rooney CM. The generation and characterization of LMP2-specific CTLs for use as adoptive transfer from patients with relapsed EBV-positive Hodgkin disease. J Immunother 2005; 27:317-27. [PMID: 15235393 DOI: 10.1097/00002371-200407000-00008] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cellular adoptive immunotherapy for virus-associated malignant disease is an attractive strategy, since viral antigens provide targets for specific T lymphocytes. In Epstein-Barr virus (EBV)-positive Hodgkin disease (HD), a limited number of EBV-encoded antigens such as the latent membrane antigens (LMP) 1 and 2 are expressed on the malignant Reed-Sternberg cells. The authors aimed to generate cytotoxic T lymphocytes (CTLs) from patients with relapsed HD by specifically targeting LMP2A. Patients with relapsed HD have highly immunosuppressive tumors and have been heavily pretreated with cytotoxic agents. As a result, monocytes and lymphocytes are numerically reduced and functionally impaired. Approaches using dendritic cells (DCs) as the sole antigen-presenting cell to expand LMP2-specific CTL lines in vitro have proved impractical. The authors now show how small amounts of patient peripheral blood can be used to produce DCs expressing LMP2 after Ad5F35 transduction, and how an initial reactivation of LMP2-specific CTLs can be followed by stimulation with lymphoblastoid cell lines overexpressing LMP2 from the same vector. Large numbers of LMP2-specific cytotoxic lymphocytes are produced that contain both CD4+ and CD8+ T cells (favoring long-term persistence in vivo) and recognize multiple LMP2 epitopes (minimizing the risk of tumor antigen loss variants). This approach is being used in a current clinical trial.
Collapse
Affiliation(s)
- Catherine M Bollard
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Lu S, Wieder E, Komanduri K, Ma Q, Molldrem JJ. Vaccines in leukemia. ADVANCES IN PHARMACOLOGY 2004; 51:255-70. [PMID: 15464913 DOI: 10.1016/s1054-3589(04)51011-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Affiliation(s)
- Sijie Lu
- The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
31
|
Burns DM, Crawford DH. Epstein–Barr virus-specific cytotoxic T-lymphocytes for adoptive immunotherapy of post-transplant lymphoproliferative disease. Blood Rev 2004; 18:193-209. [PMID: 15183903 DOI: 10.1016/j.blre.2003.12.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Post-transplant lymphoproliferative disease (PTLD) refers to a collection of clinically and pathologically diverse tumours associated with iatrogenic immunosuppression following transplantation. In most cases, tumourigenesis results from a deficit in Epstein-Barr virus (EBV)-specific cytotoxic T-lymphocyte (CTL) activity that leads to uncontrolled EBV-driven outgrowth of latently infected B-lymphocytes. Conventional treatment for PTLD typically involves a reduction in immunosuppression, but this approach is frequently unsuccessful and mortality remains high. An alternative, adoptive immunotherapy, involving the administration of EBV-specific CTLs cultured in vitro has been developed with the aim of selectively reconstituting EBV-directed immunity and effecting targeted tumour destruction. This approach has been the subject of several clinical studies, and these provide encouraging evidence of its clinical efficacy. This review presents an overview of the pathogenesis of PTLD and examines current progress in the use of adoptive immunotherapy for its treatment.
Collapse
Affiliation(s)
- David M Burns
- Basic and Clinical Virology Laboratory, Section of Medical Microbiology, University of Edinburgh, R(D)VS, Summerhall, Edinburgh EH9 1QH, UK.
| | | |
Collapse
|
32
|
Abstract
Epstein-Barr virus (EBV) is associated with a number of human malignancies. The cells of these tumours express a range of EBV latent cycle gene products that have the potential to be exploited as targets for T cell-mediated immunological therapies. Considerable progress has been made in developing adoptive T cell transfer for EBV-associated post-transplant lymphoproliferative disease (PTLD) and clinical experience clearly demonstrates that EBV-specific T cell responses can be used to treat this EBV-associated malignancy. Adoptive T cell therapies for other EBV-associated malignancies are less advanced, although encouraging data are starting to emerge. Adoptive T cell transfer, however, does require significant levels of specialist laboratory support. Large-scale treatment of patients in geographical areas with a high prevalence of EBV-associated malignancy is likely to require the development of therapeutic vaccination strategies, a number of which are in development at present. Although it remains to be seen whether long-lasting sterilising immunity to EBV could be achieved, an alternative vaccine-based approach would be to develop a prophylactic vaccine to protect against primary EBV infection.
Collapse
Affiliation(s)
- Graham S Taylor
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Vincent Drive, Birmingham B15 2TT, UK
| |
Collapse
|
33
|
Lucas KG, Salzman D, Garcia A, Sun Q. Adoptive immunotherapy with allogeneic Epstein-Barr virus (EBV)-specific cytotoxic T-lymphocytes for recurrent, EBV-positive Hodgkin disease. Cancer 2004; 100:1892-901. [PMID: 15112270 DOI: 10.1002/cncr.20188] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND It has been shown that adoptive immunotherapy with Epstein-Barr virus (EBV)-specific cytotoxic T-lymphocytes (CTL) is effective for the treatment of EBV-induced lymphoproliferative disease in stem cell transplantation recipients and organ transplantation recipients. The role of EBV CTL in other tumors for which this virus has been implicated in pathogenesis, such as EBV-positive Hodgkin disease (HD), has not been demonstrated clearly. METHODS To investigate the antitumor effects and toxicity of allogeneic EBV CTL in EBV-positive HD, the authors initiated a pilot trial in which EBV CTL were cultured from allogeneic, partially human leukocyte antigen-matched donors and were infused into patients who had therapy-refractory disease. The first cohort of 3 patients (Cohort I) received 3 separate infusions of EBV CTL (5.0 x 10(6) EBV CTL/kg per dose), and the second cohort (Cohort II) received 30 mg/m(2) per day of fludarabine for 3 days followed by a single CTL infusion (1.5 x 10(7) EBV CTL/kg). RESULTS All three patients in Cohort I had decreases in measurable disease after EBV CTL infusions, and one of those patients was without evidence of disease 22 months after infusion. Two of 3 patients in Cohort II had decreases in measurable disease, although it was not determined whether those decreases were related to fludarabine or to CTL, and 1 patient in Cohort II had 7 months without disease progression. Unlike the patients in Cohort I, fludarabine recipients did not have increases in antidonor CTL responses. Donor cells could not be detected in any of the CTL recipients. CONCLUSIONS Adoptive immunotherapy with allogeneic EBV CTL was safe for patients with recurrent, refractory, EBV-positive HD; and clinical responses may be observed without the establishment of detectable donor lymphoid chimerism.
Collapse
Affiliation(s)
- Kenneth G Lucas
- Bone Marrow Transplantation Program, Departments of Pediatrics and Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| | | | | | | |
Collapse
|
34
|
Mapara MY, Sykes M. Tolerance and cancer: mechanisms of tumor evasion and strategies for breaking tolerance. J Clin Oncol 2004; 22:1136-51. [PMID: 15020616 DOI: 10.1200/jco.2004.10.041] [Citation(s) in RCA: 414] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The development of malignant disease might be seen as a failure of immune surveillance. However, not all tumors are naturally immunogenic, and even among those that are immunogenic, the uncontrolled rapid growth of a tumor may sometimes out-run a robust immune response. Nevertheless, recent evidence suggests that mechanisms of tolerance that normally exist to prevent autoimmune disease may also preclude the development of an adequate antitumor response and that tumors themselves have the ability to thwart the development of effective immune responses against their antigens. A major challenge has been to develop approaches to breaking this tolerance in tumor-bearing hosts, and recent advances in our understanding of antigen presentation and tolerance have led to some promising strategies. An alternative approach is to use T cells from nontumor-bearing, allogeneic hosts in the form of lymphocyte infusions, with or without hematopoietic cell transplantation. Immunotherapy may occur in this setting via the response of nontolerant, tumor antigen-specific T cells from nontumor-bearing hosts or via the powerful destructive effect of an alloresponse directed against antigens shared by malignant cells in the recipient. Approaches to exploiting this beneficial effect without the deleterious consequence of graft-versus-host disease in allogeneic hematopoietic cell recipients are discussed.
Collapse
Affiliation(s)
- Markus Y Mapara
- Department of Hematology and Oncology, University Medical Center Charité, Campus Virchow Klinikum, Humboldt University Berlin, Germany
| | | |
Collapse
|
35
|
Abstract
Survivors of Hodgkin's lymphoma (HL) frequently have many years to experience the long-term toxicities of combined modality therapies. Also, a significant proportion of HL patients will relapse or have refractory disease, and less than half of these patients will respond to current salvage strategies. 30-50% of HL cases are Epstein-Barr virus associated (EBV-positive HL). The virus is localized to the malignant cells and is clonal. EBV-positive HL is more frequent in childhood, in older adults (>45 years) and in mixed cellularity cases. The survival of EBV-positive HL in the elderly and the immunosuppressed is particularly poor. Despite improvements in our understanding of EBV-positive HL, the true contribution of EBV to the pathogenesis of HL remains unknown. Increased knowledge of the virus' role in the basic biology of HL may generate novel therapeutic strategies for EBV-positive HL and the presence of EBV-latent antigens in the malignant HL cells may represent a target for cellular immunotherapy.
Collapse
Affiliation(s)
- Maher K Gandhi
- Department of Tumour Immunology, Division of Infectious Diseases and Immunology, Queensland Institute of Medical Research, Herston, Brisbane, Queensland, Australia.
| | | | | |
Collapse
|
36
|
Ong KW, Wilson AD, Hirst TR, Morgan AJ. The B subunit of Escherichia coli heat-labile enterotoxin enhances CD8+ cytotoxic-T-lymphocyte killing of Epstein-Barr virus-infected cell lines. J Virol 2003; 77:4298-305. [PMID: 12634387 PMCID: PMC150673 DOI: 10.1128/jvi.77.7.4298-4305.2003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Epstein-Barr virus (EBV) is associated with a number of important human cancers, including nasopharyngeal carcinoma, gastric carcinoma, and Hodgkin's lymphoma. These tumors express a viral nuclear antigen, EBV nuclear antigen 1 (EBNA1), which cannot be presented to T cells in a major histocompatibility complex class I context, and the viral latent membrane proteins (LMPs). Although the LMPs are expressed in these tumors, no effective immune response is made. We report here that exposure to the cholera-like enterotoxin B subunit (EtxB) in EBV-infected lymphoblastoid cell lines (LCLs) enhances their susceptibility to killing by LMP-specific CD8(+) cytotoxic T lymphocytes (CTLs) in a HLA class I-restricted manner. CTL killing of LCLs is dramatically increased through both transporter-associated protein-dependent and -independent epitopes after EtxB treatment. The use of mutant B subunits revealed that the enhanced susceptibility of LCLs to CTL killing is dependent on the B subunit's interaction with GM(1) but not its signaling properties. These important findings could underpin the development of novel approaches to treating EBV-associated malignancies and may offer a general approach to increasing the presentation of other tumor and viral antigens.
Collapse
Affiliation(s)
- Kong-Wee Ong
- Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, United Kingdom
| | | | | | | |
Collapse
|
37
|
Gottschalk S, Edwards OL, Sili U, Huls MH, Goltsova T, Davis AR, Heslop HE, Rooney CM. Generating CTLs against the subdominant Epstein-Barr virus LMP1 antigen for the adoptive immunotherapy of EBV-associated malignancies. Blood 2003; 101:1905-12. [PMID: 12411306 DOI: 10.1182/blood-2002-05-1514] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Epstein-Barr virus (EBV)-encoded LMP1 protein is expressed in EBV-positive Hodgkin disease and is a potential target for cytotoxic T-lymphocyte (CTL) therapy. However, the LMP1-specific CTL frequency is low, and so far the generation of LMP1-specific CTLs has required T-cell cloning. The toxicity of LMP1 has prevented the use of dendritic cells (DCs) for CTL stimulation, and we reasoned that an inactive, nontoxic LMP1 mutant (DeltaLMP1) could be expressed in DCs and would enable the activation and expansion of polyclonal LMP1-specific CTLs. Recombinant adenoviral vectors expressing LMP1 or DeltaLMP1 were tested for their ability to transduce DCs. LMP1 expression was toxic within 48 hours whereas high levels of DeltaLMP1 expression were achieved with minimal toxicity. DeltaLMP1-expressing DCs were able to reactivate and expand LMP1-specific CTLs from 3 healthy EBV-seropositive donors. LMP1-specific T cells were detected by interferon-gamma (IFN-gamma) enzyme-linked immunospot assay (ELISPOT) assays using the HLA-A2-restricted LMP1 peptide, YLQQNWWTL (YLQ). YLQ-specific T cells were undetectable (less than 0.001%) in donor peripheral blood mononuclear cells (PBMCs); however, after stimulation the frequency increased to 0.5% to 3.8%. Lysis of autologous target cells by CTLs was dependent on the level of LMP1 expression. In contrast, the frequency of YLQ-specific CTLs in EBV-specific CTLs reactivated and expanded using lymphoblastoid cell lines was low and no LMP1-specific cytotoxic activity was observed. Thus, DeltaLMP1 expression in DCs is nontoxic and enables the generation of LMP1-specific CTLs for future adoptive immunotherapy protocols for patients with LMP1-positive malignancies such as EBV-positive Hodgkin disease. Targeting LMP1 in these malignancies may improve the efficacy of current adoptive immunotherapy approaches.
Collapse
MESH Headings
- Adenoviruses, Human/genetics
- Antigen Presentation
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Cytotoxicity, Immunologic
- Dendritic Cells/immunology
- Epitopes/immunology
- Epstein-Barr Virus Infections/therapy
- Epstein-Barr Virus Nuclear Antigens/genetics
- Genetic Vectors/genetics
- HLA-A2 Antigen/immunology
- Hodgkin Disease/therapy
- Hodgkin Disease/virology
- Humans
- Immunotherapy, Adoptive
- Interferon-gamma/metabolism
- Lymphocyte Activation
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Transduction, Genetic
- Tumor Virus Infections/therapy
- Vaccinia virus/genetics
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/immunology
- Viral Matrix Proteins/toxicity
Collapse
Affiliation(s)
- Stephen Gottschalk
- Center for Cell and Gene Therapy, Texas Children's Cancer Center, Departments of Pediatrics, Medicine, and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Herrmann K, Niedobitek G. Lack of evidence for an association of Epstein-Barr virus infection with breast carcinoma. Breast Cancer Res 2003; 5:R13-7. [PMID: 12559053 PMCID: PMC154138 DOI: 10.1186/bcr561] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2002] [Accepted: 11/01/2002] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Epstein-Barr virus (EBV) is a ubiquitous human gamma-herpes virus infecting more than 90% of the population worldwide. EBV is associated with certain malignancies (e.g. Burkitt lymphoma, Hodgkin lymphoma and nasopharyngeal carcinoma). Recent studies have raised the possibility that EBV may also be involved in the pathogenesis of breast carcinoma, the most common carcinoma of females. If substantiated, this finding would have major implications regarding prevention and therapy of the disease. The studies published so far have employed diverse methods, however, and the results have been controversial. METHODS Using the EBV DNA PCR, EBV DNA in situ hybridisation and in situ hybridisation for the detection of the EBV-encoded RNAs, and using immunohistochemistry for the demonstration of the EBV-encoded nuclear antigen 1, we have studied a series of 59 invasive breast carcinomas for evidence of EBV infection. RESULTS EBV-encoded RNA-specific in situ hybridisation and EBV-encoded nuclear antigen 1 immunohistochemistry were negative in all cases. Using the PCR, EBV DNA was detected in four out of 59 cases. These cases were further studied by EBV DNA in situ hybridisation, showing an absence of viral DNA from the tumour cells. CONCLUSION These results indicate that breast carcinoma is not an EBV-associated tumour.
Collapse
Affiliation(s)
- Kathrin Herrmann
- Institute for Pathology, Friedrich-Alexander University, Erlangen, Germany
| | - Gerald Niedobitek
- Institute for Pathology, Friedrich-Alexander University, Erlangen, Germany
| |
Collapse
|
39
|
Shah MH, Porcu P, Mallery SR, Caligiuri MA. AIDS-associated malignancies. CANCER CHEMOTHERAPY AND BIOLOGICAL RESPONSE MODIFIERS 2003; 21:717-46. [PMID: 15338771 DOI: 10.1016/s0921-4410(03)21034-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
40
|
Niedobitek G, Päzolt D, Teichmann M, Devergne O. Frequent expression of the Epstein-Barr virus (EBV)-induced gene, EBI3, an IL-12 p40-related cytokine, in Hodgkin and Reed-Sternberg cells. J Pathol 2002; 198:310-6. [PMID: 12375263 DOI: 10.1002/path.1217] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Epstein-Barr virus (EBV)-associated Hodgkin lymphoma (HL) and nasopharyngeal carcinoma (NPC) usually occur in patients without clinically manifest deficiencies in anti-viral immunity. In spite of expressing viral proteins, both tumours are apparently able to escape EBV-specific immunity in vivo. EBI3 is an EBV-induced cytokine homologous to the interleukin (IL)-12 p40 subunit and can heterodimerize with IL-12 p35. It has been suggested that EBI3 may function to antagonize IL-12 and to inhibit the development of a Th1 immune response. EBI3 expression has been studied in tumour entities frequently associated with EBV infection to examine if EBI3 might contribute to local modulation of the immune response. It is shown that EBI3 is strongly expressed in Hodgkin and Reed-Sternberg cells in 32 of 33 HL cases, independently of the EBV status of the tumour cells. Furthermore, EBI3 expression was detected in the epithelial tumour cells of six of 40 NPC biopsies but not in Burkitt lymphomas. The results suggest that EBI3 may be an additional component of the repertoire employed by Hodgkin and Reed-Sternberg cells to inhibit an effective anti-tumour or anti-viral immune response.
Collapse
Affiliation(s)
- Gerald Niedobitek
- Institute for Pathology, Friedrich-Alexander-University, Erlangen, Germany.
| | | | | | | |
Collapse
|
41
|
Sun Q, Burton R, Reddy V, Lucas KG. Safety of allogeneic Epstein-Barr virus (EBV)-specific cytotoxic T lymphocytes for patients with refractory EBV-related lymphoma. Br J Haematol 2002; 118:799-808. [PMID: 12181048 DOI: 10.1046/j.1365-2141.2002.03683.x] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Epstein-Barr virus (EBV) causes lymphomas in immunocompromised individuals such as recipients of stem cell or organ transplants and patients with acquired immunodeficiency syndrome (AIDS). EBV has also been detected in the Reed-Sternberg cells of approximately 50% of all cases of Hodgkin's disease (HD). The purpose of this study was to examine the safety, and the clinical and immunological effects of infusing allogeneic EBV-specific cytotoxic T lymphocytes (CTL) for patients with refractory EBV-positive malignancies. In this pilot study, we have treated four patients with EBV-related lymphoma using allogeneic EBV-specific CTL. Two patients received EBV-specific CTL derived from partially human leucocyte antigen (HLA)-matched donors and the other two from HLA-matched siblings. No complications were observed as a result of the CTL infusions and all patients showed increased levels of EBV-specific CTL precursors (CTLp) post infusion. Of the two organ transplant patients, one had refractory disease and has sustained a complete remission following the T-cell infusions. The second has also been disease free since T-cell infusions, although the efficacy cannot be definitively attributed to CTL therapy because this patient received local radiation therapy prior to immunotherapy. A patient with AIDS-related, EBV-positive lymphoma had disease progression following CTL infusions. One HD patient received HLA 4/6 matched T cells from an unrelated donor and showed a decrease in the size of affected lymph nodes and resolution of B-symptoms post infusion. In conclusion, adoptive immunotherapy with allogeneic EBV-specific CTL is safe and may have efficacy in patients with high-risk or refractory EBV-related tumours.
Collapse
Affiliation(s)
- Qi Sun
- Department of Medicine, University of Alabama at Birmingham, 35294, USA
| | | | | | | |
Collapse
|
42
|
Meij P, Leen A, Rickinson AB, Verkoeijen S, Vervoort MBHJ, Bloemena E, Middeldorp JM. Identification and prevalence of CD8(+) T-cell responses directed against Epstein-Barr virus-encoded latent membrane protein 1 and latent membrane protein 2. Int J Cancer 2002; 99:93-9. [PMID: 11948498 DOI: 10.1002/ijc.10309] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Epstein-Barr virus (EBV) is associated with several human malignancies that each show different viral gene expression profiles. In malignancies such as Hodgkin's disease and nasopharyngeal carcinoma only Epstein-Barr nuclear antigen 1 (EBNA1) and varying levels of latent membrane proteins 1 and 2 (LMP1 and -2) are expressed. Since endogenously expressed EBNA1 is protected from CTL recognition, LMP1 and LMP2 are the most likely target antigens for anti-tumor immunotherapy. Therefore, we sought to identify in a systematic way CD8(+) T-cell responses directed against eptitopes derived from LMP1 and LMP2. Using IFNgamma-ELISPOT assays of interferon-gamma release, peripheral blood mononuclear cells (PBMC) of healthy donors were screened with peptide panels (15 mer overlapping by 10) spanning the LMP1 and LMP2 sequences of the prototype EBV strain B95.8. When positive responses were found, CD4(+) or CD8(+) T cells were depleted from donor PBMC to determine the origin of the responder population. We detected CD8(+) T-cell responses to LMP1 in 9/50(18%) donors and to LMP2 in 15/28 (54%) donors. In addition to the already described epitopes, 3 new LMP1- and 5 new LMP2-derived CD8(+) epitopes were identified. In most donors LMP1- and LMP2-specific CD8(+) precursor frequencies were low compared with precursors against immunodominant EBV epitopes from latent (EBNA3A, -3B and -3C) and lytic cycle antigens. These results demonstrate that CD8(+) memory T cell responses to LMP1 and especially to LMP2 do exist in Caucasians, albeit at low levels and could potentially be exploited for therapeutic use.
Collapse
Affiliation(s)
- Pauline Meij
- Department of Pathology, VU Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
43
|
Sjöberg J, Andersson M, Garcia C, Palucka KA, Björkholm M, Porwit-MacDonald A, Pisa P. Expression of the signal transduction molecule zeta in peripheral and tumour-associated lymphocytes in Hodgkin's disease in relation to the Epstein-Barr virus status of the tumour cells. Br J Haematol 2002; 116:765-73. [PMID: 11886379 DOI: 10.1046/j.0007-1048.2002.03342.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We investigated whether the described immune evasion of Epstein-Barr virus (EBV)-infected malignant Hodgkin and Reed-Sternberg (HRS) cells in Hodgkin's disease (HD) is paralleled by a disturbed expression of the signal transduction molecule zeta associated with CD3 and CD16 in tumour-associated T lymphocytes (TAL). Flow cytometric analysis revealed a significantly lower zeta expression in CD3+/4+, CD3+/8+ and CD16+ patient peripheral blood lymphocytes (PBL; n = 10) compared with normal donor PBLs (n = 11). When patient PBLs were compared with the corresponding TAL, the latter showed a significantly higher (CD3+/4+) or equal (CD3+/8+) zeta expression. The EBV status of the tumours did not correlate with zeta expression in the TAL. Immunohistochemical staining revealed zeta-positive lymphocytes among the adjacent bystander cells of the HRS cells in all analysed tumours (n = 8), irrespective of tumour EBV status. In conclusion, these results do not support downregulation of zeta in TAL as a critical mechanism contributing specifically to the immune escape of EBV+ HRS cells.
Collapse
Affiliation(s)
- Jan Sjöberg
- Department of Medicine, Pathology and Oncology, Radiumhemmet, Karolinska Hospital, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | |
Collapse
|
44
|
Gan YJ, Razzouk BI, Su T, Sixbey JW. A defective, rearranged Epstein-Barr virus genome in EBER-negative and EBER-positive Hodgkin's disease. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 160:781-6. [PMID: 11891176 PMCID: PMC1867161 DOI: 10.1016/s0002-9440(10)64900-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A ubiquitous herpesvirus that establishes life-long infection, the Epstein-Barr virus (EBV) has yielded little insight into how a single agent in general accord with its host can produce diverse pathologies ranging from oral hairy leukoplakia to nasopharyngeal carcinoma, from infectious mononucleosis to Hodgkin's disease (HD) and Burkitt's lymphoma. Its pathogenesis is further confounded by the less than total association of virus with histologically similar tumors. In other viral systems, defective (interfering) viral genomes are known to modulate outcome of infection, with either ameliorating or intensifying effects on disease processes initiated by prototype strains. To ascertain whether defective EBV genomes are present in HD, we examined paraffin-embedded tissue from 56 HD cases whose EBV status was first determined by cytohybridization for nonpolyadenylated EBV RNAs (EBERs). Using both standard polymerase chain reaction (PCR) and PCR in situ hybridization, we successfully amplified sequences that span abnormally juxtaposed BamHI W and Z fragments characteristic of defective heterogeneous (het) EBV DNA from 10 of 32 (31%) EBER-positive tumors. Of 24 EBER-negative HD, 8 yielded PCR products indicating presence of het EBV DNA. Two of these contained defective EBV in the apparent absence of the prototype virus. Of the 42 tumors analyzed for defective EBV by both PCR techniques, there was concordance of results in 38 (90%). Detection of defective EBV genomes with the potential to disrupt viral gene regulation suggests one mechanism for pathogenic diversity that may also account for loss of prototypic EBV from individual tumor cells.
Collapse
Affiliation(s)
- Yan-Jun Gan
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, USA
| | | | | | | |
Collapse
|
45
|
Oelmann E, Herbst H, Zühlsdorf M, Albrecht O, Nolte A, Schmitmann C, Manzke O, Diehl V, Stein H, Berdel WE. Tissue inhibitor of metalloproteinases 1 is an autocrine and paracrine survival factor, with additional immune-regulatory functions, expressed by Hodgkin/Reed-Sternberg cells. Blood 2002; 99:258-67. [PMID: 11756180 DOI: 10.1182/blood.v99.1.258] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Tissue inhibitor of metalloproteinases (TIMP)-1 and TIMP-2 are proteins with proteinase-inhibiting and cytokine properties. TIMP-1 is active primarily in B cells and B-cell lymphomas, whereas TIMP-2 expression is restricted to T cells. The expression of TIMP-1 and TIMP-2 in lymph nodes from patients with Hodgkin disease (HD) and in Hodgkin-derived cell lines was investigated. In situ hybridization showed TIMP-1 RNA expression in 3% to 80% of Hodgkin/Reed-Sternberg (H/R-S) cells from 14 of 15 patients, with results in one patient being at the lowest detection limit; no expression of TIMP-2 in H/R-S cells; and only weak expression of TIMP-2 in reactive lymphoid tissue. Production of TIMP-1 protein by H/R-S cells was accordingly found on immunohistochemical analysis of lymph nodes from patients with HD. There was only low expression of matrix metalloproteinase (MMP)-2, which is mainly inhibited by TIMP-2; no expression of MMP-1 and MMP-3 in reactive lymphoid tissue; and no expression of these MMPs in H/R-S cells. Thus, TIMP-1 expression in lymph nodes was not correlated with metalloproteinase expression. Five of 7 Hodgkin-derived cell lines expressed TIMP-1 at the protein level. Only one of these cell lines expressed TIMP-2, at the lowest detection limit. TIMP-1 levels in plasma from patients with HD were within the same range as those in plasma from healthy controls. Recombinant human TIMP-1 inhibited induced cell death in Hodgkin-derived cell lines in vitro. TIMP-1 and TIMP-2 inhibited T-cell cytotoxicity against autologous cells presenting tumor-associated antigens and in allogeneic mixed lymphocyte cultures. Thus, TIMP-1, aside from its role in proteinase equilibrium, is an autocrine and paracrine survival factor for H/R-S cells and an immunosuppressive protein expressed in Hodgkin lymphomas.
Collapse
Affiliation(s)
- Elisabeth Oelmann
- Department of Medicine (Hematology/Oncology), Westfaelische Wilhelms Universitaet Muenster, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Magrath I. Editorial comment on detection of Epstein-Barr virus DNA in peripheral blood of paediatric patients with Hodgkin's disease by real-time polymerase chain reaction by Wagner and colleagues. Eur J Cancer 2001; 37:1812-5. [PMID: 11576832 DOI: 10.1016/s0959-8049(01)00221-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
47
|
Gahn B, Siller-Lopez F, Pirooz AD, Yvon E, Gottschalk S, Longnecker R, Brenner MK, Heslop HE, Aguilar-Cordova E, Rooney CM. Adenoviral gene transfer into dendritic cells efficiently amplifies the immune response to LMP2A antigen: a potential treatment strategy for Epstein-Barr virus--positive Hodgkin's lymphoma. Int J Cancer 2001; 93:706-13. [PMID: 11477583 DOI: 10.1002/ijc.1396] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The EBV-encoded LMP2A protein is consistently expressed in EBV(+) Hodgkin's lymphoma and can be targeted by CTLs. CTLs stimulated conventionally by LCLs have little activity against LMP2A(+) target cells. Here, we describe an alternative approach, based on the in vitro stimulation of CTLs with DCs genetically modified with 2 E1/E3-deleted recombinant adenoviruses, AdGFPLMP2A, encoding a fusion gene of GFP and LMP2A, and AdLMP2A, encoding LMP2A only. Transduction of DCs with AdGFPLMP2A at MOI 1,000 resulted in LMP2A expression in up to 88% of DCs. LMP2A protein was expressed in 40% of DCs transduced with AdLMP2A at an MOI of 100. Higher MOI resulted in DC death. CTL lines activated by transduced DCs had a higher frequency of LMP2A tetramer-specific CTLs than CTL lines activated by LCLs. CTLs stimulated with transduced DCs lysed both autologous fibroblasts infected with vaccinia virus LMP2A (FBvaccLMP2A) and autologous LCLs, which express LMP2A at lower levels. In contrast, CTLs generated from the same donors by stimulation with autologous LCLs showed minimal lysis of FBvaccLMP2A. Moreover, 1 donor who did not respond to LMP2A when CTLs were stimulated with LCLs became a responder when LMP2A was expressed by transduced DCs. Hence, recombinant adenoviruses encoding LMP2A effectively transduce DCs and direct the generation of LMP2A-specific CTLs. This approach will be a potent strategy in Hodgkin's lymphoma immunotherapy.
Collapse
Affiliation(s)
- B Gahn
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Niedobitek G, Meru N, Delecluse HJ. Epstein-Barr virus infection and human malignancies. Int J Exp Pathol 2001. [PMID: 11488990 DOI: 10.1111/j.1365-2613.2001.iep190.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The Epstein-Barr virus (EBV) is a herpes virus which establishes a life-long persistent infection in over 90% of the human adult population world-wide. Based on its association with a variety of lymphoid and epithelial malignancies, EBV has been classified as a group 1 carcinogen by the International Agency for Research on Cancer. In this article we discuss the evidence supporting an aetiological role for EBV in the pathogenesis of human tumours. The biology of EBV infection will be described with special emphasis on viral transforming gene products. A brief survey of EBV-associated tumours is followed by a discussion of specific problems. Evidence is presented which suggests that failures of the EBV-specific immunity may play a role in the pathogenesis of EBV-associated tumours also in patients without clinically manifest immunodeficiencies. Finally, the timing of EBV infection in the pathogenesis of virus-associated malignancies is discussed. There is good evidence that EBV infection precedes expansion of the malignant cell populations in some virus-associated tumours. However, this is clearly not always the case and for some of these tumours there are indications that clonal genetic alterations may occur prior to EBV infection. Thus, whilst there is good evidence to suggest that EBV is a human carcinogen, its precise role(s) in the development of virus-associated human tumours requires clarification.
Collapse
Affiliation(s)
- G Niedobitek
- Pathologisches Institut, Friedrich-Alexander-Universität, Krankenhausstr. 8-10, 91054 Erlangen, Germany. gerald.niedobitek @patho.imed.uni-erlangen.de
| | | | | |
Collapse
|
49
|
zum Büschenfelde CM, Metzger J, Hermann C, Nicklisch N, Peschel C, Bernhard H. The generation of both T killer and Th cell clones specific for the tumor-associated antigen HER2 using retrovirally transduced dendritic cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:1712-9. [PMID: 11466395 DOI: 10.4049/jimmunol.167.3.1712] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Induction of antitumor immunity involves the presence of both CD8(+) CTLs and CD4(+) Th cells specific for tumor-associated Ags. Attempts to eradicate cancer by adoptive T cell transfer have been limited due to the difficulty of generating T cells with defined Ag specificity. The current study focuses on the generation of CTL and Th cells against the tumor-associated Ag HER2 using autologous dendritic cells (DC) derived from CD34(+) hematopoietic progenitor cells which have been retrovirally transduced with the human epidermal growth factor receptor 2 (HER2) gene. HER2-transduced DC elicited HER2-specific CD8(+) CTL that lyse HER2-overexpressing tumor cells in context of distinct HLA class I alleles. The induction of both HLA-A2 and -A3-restricted HER2-specific CTL was verified on a clonal level. In addition, retrovirally transduced DC induced CD4(+) Th1 cells recognizing HER2 in context with HLA class II. HLA-DR-restricted CD4(+) T cells were cloned that released IFN-gamma upon stimulation with DC pulsed with the recombinant protein of the extracellular domain of HER2. These data indicate that retrovirally transduced DC expressing the HER2 molecule present multiple peptide epitopes and subsequently elicit HER2-specific CTL and Th1 cells. The method of stimulating HER2-specific CD8(+) and CD4(+) T cells with retrovirally transduced DC was successfully implemented for generating HER2-specific CTL and Th1 clones from a patient with HER2-overexpressing breast cancer. The ability to generate and expand HER2-specific, HLA-restricted CTL and Th1 clones in vitro facilitates the development of immunotherapy regimens, in particular the adoptive transfer of both autologous HER2-specific T cell clones in patients with HER2-overexpressing tumors without the requirement of defining immunogenic peptides.
Collapse
MESH Headings
- Alleles
- Antigen Presentation/genetics
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/immunology
- Cell Line, Transformed
- Clone Cells
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/metabolism
- Female
- Gene Expression Regulation, Neoplastic/immunology
- Genes, erbB-2/immunology
- Histocompatibility Antigens Class I/genetics
- Humans
- Hybridomas
- Lymphocyte Activation/genetics
- Middle Aged
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Retroviridae/genetics
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- Th1 Cells/immunology
- Transduction, Genetic
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- C M zum Büschenfelde
- Department of Hematology/Oncology, Technical University of Munich, Ismaningerstrasse 22, 81664 Munich, Germany
| | | | | | | | | | | |
Collapse
|
50
|
Beck A, Päzolt D, Grabenbauer GG, Nicholls JM, Herbst H, Young LS, Niedobitek G. Expression of cytokine and chemokine genes in Epstein-Barr virus-associated nasopharyngeal carcinoma: comparison with Hodgkin's disease. J Pathol 2001; 194:145-51. [PMID: 11400141 DOI: 10.1002/path.867] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Nasopharyngeal carcinoma (NPC) and Hodgkin's disease (HD) are characterized by their association with Epstein-Barr virus (EBV) and the presence of an intense lymphoid stroma, consisting of T lymphocytes and other reactive cells. In both entities, the tumour cells express viral proteins known to provide target epitopes for cytotoxic T-cells (CTLs), yet in vivo, the tumour cells appear to escape CTL recognition. A comparative in situ hybridization study of cytokine and chemokine gene expression in NPC and HD has been undertaken, focusing on cytokines which are known to be inducible by EBV in vitro. Hodgkin and Reed-Sternberg (HRS) cells expressed interleukin (IL)-6, IL-8, and IL-10, and the thymus and activation regulated chemokine (TARC) in 15/22, 0/22, 5/22, and 16/21 cases, respectively. In NPC, the epithelial tumour cells showed expression of IL-6 in 3/43 cases and of IL-8 in 2/40 cases. There was no detectable expression of IL-10 and TARC in these cases. These data confirm that HRS cells frequently express cytokine and chemokine genes and suggest that this may enable HRS cells to modulate the immune response in their microenvironment and to escape CTL detection. In contrast, NPC tumour cells show only rare expression of IL-6 and IL-8 and no detectable expression of IL-10 and TARC. Thus, the results suggest that the mechanisms employed by the EBV-positive tumour cells to escape immune recognition and destruction differ between HD and NPC.
Collapse
Affiliation(s)
- A Beck
- Institute for Pathology, Friedrich-Alexander-University, 91054 Erlangen, Germany
| | | | | | | | | | | | | |
Collapse
|