1
|
Escalante GM, Reidel IG, Mutsvunguma LZ, Cua S, Tello BA, Rodriguez E, Farelo MA, Zimmerman C, Muniraju M, Li H, Govindan AN, Axthelm MK, Wong SW, Ogembo JG. Multivalent MVA-vectored vaccine elicits EBV neutralizing antibodies in rhesus macaques that reduce EBV infection in humanized mice. Front Immunol 2024; 15:1445209. [PMID: 39346922 PMCID: PMC11427267 DOI: 10.3389/fimmu.2024.1445209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
Introduction Epstein-Barr virus (EBV) is an oncogenic human herpesvirus associated with ~350,000 cases of lymphoid and epithelial malignancies every year, and is etiologically linked to infectious mononucleosis and multiple sclerosis. Despite four decades of research, no EBV vaccine candidate has yet reached licensure. Most previous vaccine attempts focused on a single viral entry glycoprotein, gp350, but recent data from clinical and pre-clinical studies, and the elucidation of viral entry mechanisms, support the inclusion of multiple entry glycoproteins in EBV vaccine design. Methods Here we generated a modified vaccinia Ankara (MVA)-vectored EBV vaccine, MVA-EBV5-2, that targets five EBV entry glycoproteins, gp350, gB, and the gp42gHgL complex. We characterized the genetic and translational stability of the vaccine, followed by immunogenicity assessment in BALB/c mice and rhesus lymphocryptovirus-negative rhesus macaques as compared to a gp350-based MVA vaccine. Finally, we assessed the efficacy of MVA-EBV5-2-immune rhesus serum at preventing EBV infection in human CD34+ hematopoietic stem cell-reconstituted NSG mice, under two EBV challenge doses. Results The MVA-EBV5-2 vaccine was genetically and translationally stable over 10 viral passages as shown by genetic and protein expression analysis, and when administered to female and male BALB/c mice, elicited serum EBV-specific IgG of both IgG1 and IgG2a subtypes with neutralizing activity in vitro. In Raji B cells, this neutralizing activity outperformed that of serum from mice immunized with a monovalent MVA-vectored gp350 vaccine. Similarly, MVA-EBV5-2 elicited EBV-specific IgG in rhesus macaques that were detected in both serum and saliva of immunized animals, with serum antibodies demonstrating neutralizing activity in vitro that outperformed serum from MVA-gp350-immunized macaques. Finally, pre-treatment with serum from MVA-EBV5-2-immunized macaques resulted in fewer EBV-infected mice in the two challenge experiments than pretreatment with serum from pre-immune macaques or macaques immunized with the monovalent gp350-based vaccine. Discussion These results support the inclusion of multiple entry glycoproteins in EBV vaccine design and position our vaccine as a strong candidate for clinical translation.
Collapse
Affiliation(s)
- Gabriela M. Escalante
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Ivana G. Reidel
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Lorraine Z. Mutsvunguma
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Simeon Cua
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Brenda A. Tello
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Esther Rodriguez
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
- Irell & Manella Graduate School of Biological Sciences of City of Hope, Duarte, CA, United States
| | - Mafalda A. Farelo
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Cloe Zimmerman
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
- Irell & Manella Graduate School of Biological Sciences of City of Hope, Duarte, CA, United States
| | - Murali Muniraju
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - He Li
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
| | - Aparna N. Govindan
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
| | - Michael K. Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Scott W. Wong
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, United States
| | - Javier Gordon Ogembo
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| |
Collapse
|
2
|
Dasari V, McNeil LK, Beckett K, Solomon M, Ambalathingal G, Thuy TL, Panikkar A, Smith C, Steinbuck MP, Jakubowski A, Seenappa LM, Palmer E, Zhang J, Haqq CM, DeMuth PC, Khanna R. Lymph node targeted multi-epitope subunit vaccine promotes effective immunity to EBV in HLA-expressing mice. Nat Commun 2023; 14:4371. [PMID: 37553346 PMCID: PMC10409721 DOI: 10.1038/s41467-023-39770-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 06/28/2023] [Indexed: 08/10/2023] Open
Abstract
The recent emergence of a causal link between Epstein-Barr virus (EBV) and multiple sclerosis has generated considerable interest in the development of an effective vaccine against EBV. Here we describe a vaccine formulation based on a lymph node targeting Amphiphile vaccine adjuvant, Amphiphile-CpG, admixed with EBV gp350 glycoprotein and an engineered EBV polyepitope protein that includes 20 CD8+ T cell epitopes from EBV latent and lytic antigens. Potent gp350-specific IgG responses are induced in mice with titers >100,000 in Amphiphile-CpG vaccinated mice. Immunization including Amphiphile-CpG also induces high frequencies of polyfunctional gp350-specific CD4+ T cells and EBV-specific CD8+ T cells that are 2-fold greater than soluble CpG and are maintained for >7 months post immunization. This combination of broad humoral and cellular immunity against multiple viral determinants is likely to provide better protection against primary infection and control of latently infected B cells leading to protection against the development of EBV-associated diseases.
Collapse
Affiliation(s)
- Vijayendra Dasari
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia.
| | | | - Kirrilee Beckett
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia
| | - Matthew Solomon
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia
| | - George Ambalathingal
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia
| | - T Le Thuy
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia
| | - Archana Panikkar
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia
| | - Caitlyn Smith
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia
| | | | | | | | | | - Jeff Zhang
- Elicio Therapeutics, Inc, Boston, MA, USA
| | | | | | - Rajiv Khanna
- QIMR Centre for Immunotherapy and Vaccine Development, Tumour Immunology Laboratory, Infection and Inflammation Program, Berghofer Medical Research Institute, Brisbane, Australia.
| |
Collapse
|
3
|
Lambert N, El Moussaoui M, Baron F, Maquet P, Darcis G. Virus-Specific T-Cell Therapy for Viral Infections of the Central Nervous System: A Review. Viruses 2023; 15:1510. [PMID: 37515196 PMCID: PMC10383098 DOI: 10.3390/v15071510] [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: 06/08/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Opportunistic viral infections of the central nervous system represent a significant cause of morbidity and mortality among an increasing number of immunocompromised patients. Since antiviral treatments are usually poorly effective, the prognosis generally relies on the ability to achieve timely immune reconstitution. Hence, strategies aimed at reinvigorating antiviral immune activity have recently emerged. Among these, virus-specific T-cells are increasingly perceived as a principled and valuable tool to treat opportunistic viral infections. Here we briefly discuss how to develop and select virus-specific T-cells, then review their main indications in central nervous system infections, including progressive multifocal leukoencephalopathy, CMV infection, and adenovirus infection. We also discuss their potential interest in the treatment of progressive multiple sclerosis, or EBV-associated central nervous system inflammatory disease. We finish with the key future milestones of this promising treatment strategy.
Collapse
Affiliation(s)
- Nicolas Lambert
- Department of Neurology, University Hospital of Liège, 4000 Liège, Belgium
| | - Majdouline El Moussaoui
- Department of General Internal Medicine and Infectious Diseases, University Hospital of Liège, 4000 Liège, Belgium
| | - Frédéric Baron
- Department of Hematology, University Hospital of Liège, 4000 Liège, Belgium
| | - Pierre Maquet
- Department of Neurology, University Hospital of Liège, 4000 Liège, Belgium
| | - Gilles Darcis
- Department of General Internal Medicine and Infectious Diseases, University Hospital of Liège, 4000 Liège, Belgium
| |
Collapse
|
4
|
Tan H, Gong Y, Liu Y, Long J, Luo Q, Faleti OD, Lyu X. Advancing therapeutic strategies for Epstein-Barr virus-associated malignancies through lytic reactivation. Biomed Pharmacother 2023; 164:114916. [PMID: 37229802 DOI: 10.1016/j.biopha.2023.114916] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023] Open
Abstract
Epstein-Barr virus (EBV) is a widespread human herpes virus associated with lymphomas and epithelial cell cancers. It establishes two separate infection phases, latent and lytic, in the host. Upon infection of a new host cell, the virus activates several pathways, to induce the expression of lytic EBV antigens and the production of infectious virus particles. Although the carcinogenic role of latent EBV infection has been established, recent research suggests that lytic reactivation also plays a significant role in carcinogenesis. In this review, we summarize the mechanism of EBV reactivation and recent findings about the role of viral lytic antigens in tumor formation. In addition, we discuss the treatment of EBV-associated tumors with lytic activators and the targets that may be therapeutically effective in the future.
Collapse
Affiliation(s)
- Haiqi Tan
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China
| | - Yibing Gong
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China
| | - Yi Liu
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China
| | - Jingyi Long
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China
| | - Qingshuang Luo
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China
| | - Oluwasijibomi Damola Faleti
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, 999000, Hong Kong Special Administrative Region of China
| | - Xiaoming Lyu
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China.
| |
Collapse
|
5
|
Smith C, Khanna R. Adoptive T-cell therapy targeting Epstein-Barr virus as a treatment for multiple sclerosis. Clin Transl Immunology 2023; 12:e1444. [PMID: 36960148 PMCID: PMC10028422 DOI: 10.1002/cti2.1444] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/23/2023] Open
Abstract
Emergence of a definitive link between Epstein-Barr virus (EBV) and multiple sclerosis has provided an impetus to develop immune-based therapies to target EBV-infected B cells. Initial studies with autologous EBV-specific T-cell therapy demonstrated that this therapy is safe with minimal side effects and more importantly multiple patients showed both symptomatic and objective neurological improvements including improved quality of life, reduction of fatigue and reduced intrathecal IgG production. These observations have been successfully extended to an 'off-the-shelf' allogeneic EBV-specific T-cell therapy manufactured using peripheral blood lymphocytes of healthy seropositive individuals. This adoptive immunotherapy has also been shown to be safe with encouraging clinical responses. Allogeneic EBV T-cell therapy overcomes some of the limitations of autologous therapy and can be rapidly delivered to patients with improved therapeutic potential.
Collapse
Affiliation(s)
- Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, Infection and Inflammation ProgramQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Rajiv Khanna
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, Infection and Inflammation ProgramQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| |
Collapse
|
6
|
Venkataraman T, Valencia C, Mangino M, Morgenlander W, Clipman SJ, Liechti T, Valencia A, Christofidou P, Spector T, Roederer M, Duggal P, Larman HB. Analysis of antibody binding specificities in twin and SNP-genotyped cohorts reveals that antiviral antibody epitope selection is a heritable trait. Immunity 2022; 55:174-184.e5. [PMID: 35021055 PMCID: PMC8852220 DOI: 10.1016/j.immuni.2021.12.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/19/2021] [Accepted: 12/07/2021] [Indexed: 01/13/2023]
Abstract
Human immune responses to viral infections are highly variable, but the genetic factors that contribute to this variability are not well characterized. We used VirScan, a high-throughput epitope scanning technology, to analyze pan-viral antibody reactivity profiles of twins and SNP-genotyped individuals. Using these data, we determined the heritability and genomic loci associated with antibody epitope selection, response breadth, and control of Epstein-Barr virus (EBV) viral load. 107 EBV peptide reactivities were heritable and at least two Epstein-Barr nuclear antigen 2 (EBNA-2) reactivities were associated with variants in the MHC class II locus. We identified an EBV serosignature that predicted viral load in peripheral blood mononuclear cells and was associated with variants in the MHC class I locus. Our study illustrates the utility of epitope profiling to investigate the genetics of pathogen immunity, reports heritable features of the antibody response to viruses, and identifies specific HLA loci important for EBV epitope selection.
Collapse
Affiliation(s)
- Thiagarajan Venkataraman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Cristian Valencia
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Massimo Mangino
- Department of Twin Research & Genetic Epidemiology, King’s College of London, London, UK,NIHR Biomedical Research Centre at Guy’s and St Thomas’ Foundation Trust, London SE1 9RT, UK
| | - William Morgenlander
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Steven J. Clipman
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Thomas Liechti
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Ana Valencia
- School of Medicine, Universidad Pontificia Bolivariana, Medellín, Colombia
| | - Paraskevi Christofidou
- Department of Twin Research & Genetic Epidemiology, King’s College of London, London, UK
| | - Tim Spector
- Department of Twin Research & Genetic Epidemiology, King’s College of London, London, UK
| | - Mario Roederer
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Priya Duggal
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, USA,Lead contact,Correspondence: (H.B.L)
| |
Collapse
|
7
|
Sun C, Chen XC, Kang YF, Zeng MS. The Status and Prospects of Epstein-Barr Virus Prophylactic Vaccine Development. Front Immunol 2021; 12:677027. [PMID: 34168649 PMCID: PMC8218244 DOI: 10.3389/fimmu.2021.677027] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 05/20/2021] [Indexed: 12/30/2022] Open
Abstract
Epstein–Barr virus (EBV) is a human herpesvirus that is common among the global population, causing an enormous disease burden. EBV can directly cause infectious mononucleosis and is also associated with various malignancies and autoimmune diseases. In order to prevent primary infection and subsequent chronic disease, efforts have been made to develop a prophylactic vaccine against EBV in recent years, but there is still no vaccine in clinical use. The outbreak of the COVID-19 pandemic and the global cooperation in vaccine development against SARS-CoV-2 provide insights for next-generation antiviral vaccine design and opportunities for developing an effective prophylactic EBV vaccine. With improvements in antigen selection, vaccine platforms, formulation and evaluation systems, novel vaccines against EBV are expected to elicit dual protection against infection of both B lymphocytes and epithelial cells. This would provide sustainable immunity against EBV-associated malignancies, finally enabling the control of worldwide EBV infection and management of EBV-associated diseases.
Collapse
Affiliation(s)
- Cong Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Xin-Chun Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Yin-Feng Kang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
Abstract
Glycoprotein B (gB) is an essential fusion protein for the Epstein-Barr virus (EBV) infection of both B cells and epithelial cells and is thus a promising target antigen for a prophylactic vaccine to prevent or reduce EBV-associated disease. T cell responses play key roles in the control of persistent EBV infection and in the efficacy of a vaccine. However, to date, T cell responses to gB have been characterized for only a limited number of human leukocyte antigen (HLA) alleles. Here, we screened gB T cell epitopes in 23 healthy EBV carriers and ten patients with nasopharyngeal cancer (NPC) using a peptide library spanning the entire gB sequence. We identified twelve novel epitopes in the context of seven new HLA restrictions that are common in Asian populations. Two epitopes, gB214-223 and gB840-849, restricted by HLA-B*58:01 and B*38:02, respectively, elicited specific CD8+ T cell responses to inhibit EBV-driven B cell transformation. Interestingly, gB-specific CD8+ T cells were more frequent in healthy viral carriers with EBV reactivation than in those without EBV reactivation, indicating that EBV reactivation in vivo stimulates both humoral (VCA-gp125-IgA) and cellular responses to gB. We further found that most gB epitopes are conserved among different EBV strains. Our study broadens the diversity and HLA restrictions of gB epitopes and suggests that gB is a common target of T cell responses in healthy viral carriers with EBV reactivation. In particular, the precisely mapped and conserved gB epitopes provide valuable information for prophylactic vaccine development.ImportanceT cells are crucial for the control of persistent EBV infection and the development of EBV-associated diseases. The EBV gB protein is essential for virus entry into B cells and epithelial cells and is thus a target antigen for vaccine development. Understanding T cell responses to gB is important for subunit vaccine design. Herein, we comprehensively characterized T cell responses to full-length gB. Our results expand the available gB epitopes and HLA restrictions, particularly those common in Asian populations. Furthermore, we showed that gB-specific CD8+ T cells inhibit B cell transformation ex vivo and that gB-specific CD8+ T cell responses in vivo may be associated with intermittent EBV reactivation in asymptomatic viral carriers. These gB epitopes are highly conserved among geographically separated EBV strains. Precisely mapped and conserved T cell epitopes may contribute to immune monitoring and to the development of a gB subunit vaccine.
Collapse
|
9
|
Functional Differences Between EBV- and CMV-Specific CD8 + T cells Demonstrate Heterogeneity of T cell Dysfunction in CLL. Hemasphere 2020; 4:e337. [PMID: 32309780 PMCID: PMC7162091 DOI: 10.1097/hs9.0000000000000337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/18/2019] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
Acquired T cell dysfunction is a hallmark of chronic lymphocytic leukemia (CLL), and is linked to an increased risk of infections, but also reduced immune surveillance and disappointing responses to autologous T cell-based immunotherapy. The mechanisms of T cell dysfunction in CLL are not well understood. Studying immunity against chronic viruses allows for detailed analysis of the effect of CLL on T cells chronically exposed to a specific antigen. Cytomegalovirus (CMV) reactivations are rare in CLL, which corroborates with preserved CMV-specific T cell function. Epstein-Barr virus (EBV) is another herpesvirus that results in chronic infection, but unlike CMV, is characterized by subclinical reactivations in CLL patients. Since both herpesviruses induce strong CD8+ T cell responses, but have different clinical outcomes, studying these specific T cells may shed light on the mechanisms of CLL-induced T cell dysfunction. We first analyzed the phenotype of EBV-specific CD8+ T cells in CLL and healthy controls, and found that in CLL EBV-specific CD8+ T cells are in an advanced differentiation state with higher expression of inhibitory receptors. Secondly, CLL-derived EBV-specific CD8+ T cells show reduced cytotoxic potential, in contrast to CMV-specific T cells. Finally, we performed transcriptome analysis to visualize differential modulation by CLL of these T cell subsets. While T cell activation and differentiation genes are unaffected, in EBV-specific T cells expression of genes involved in synapse formation and T cell exhaustion is altered. Our findings on the heterogeneity of antigen specific T cell function in CLL aids in understanding immune-dysregulation in this disease.
Collapse
|
10
|
Pender MP. Hypothesis: bipolar disorder is an Epstein-Barr virus-driven chronic autoimmune disease - implications for immunotherapy. Clin Transl Immunology 2020; 9:e1116. [PMID: 32257210 PMCID: PMC7133420 DOI: 10.1002/cti2.1116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/09/2020] [Accepted: 02/10/2020] [Indexed: 01/29/2023] Open
Abstract
Bipolar disorder (BD) is a chronic disease characterised by episodes of major depression and episodes of mania or hypomania, with a worldwide prevalence of 2.4%. The cause of BD is unknown. Here, I propose the hypothesis that BD is a chronic autoimmune disease caused by Epstein–Barr virus (EBV) infection of autoreactive B cells. It is postulated that EBV‐infected autoreactive B cells accumulate in the brain where they provide costimulatory survival signals to autoreactive T cells and differentiate into plasma cells producing pathogenic autoantibodies targeting brain components such as the N‐methyl‐D‐aspartate receptor. It is also proposed that the accumulation of EBV‐infected autoreactive B cells in the brain is a consequence of a genetically determined defect in the ability of CD8+ T cells to control EBV infection. The theory is supported by studies indicating that autoimmunity, EBV infection and CD8+ T‐cell deficiency all have roles in the pathogenesis of BD. According to the hypothesis, BD should be able to be treated by EBV‐specific T‐cell therapy and to be prevented by vaccination against EBV in early childhood. Exposure to sunlight or appropriate artificial light should also be beneficial in BD by augmenting CD8+ T‐cell control of EBV infection.
Collapse
Affiliation(s)
- Michael P Pender
- Faculty of Medicine The University of Queensland Brisbane QLD Australia.,Department of Neurology Royal Brisbane and Women's Hospital Brisbane QLD Australia
| |
Collapse
|
11
|
Park SS, Cho SY, Han E, Min GJ, Park S, Yoon JH, Lee SE, Cho BS, Eom KS, Kim YJ, Lee S, Kim HJ, Min CK, Cho SG, Lee JW. Reactivation and dynamics of cytomegalovirus and Epstein-Barr virus after rabbit antithymocyte globulin and cyclosporine for aplastic anemia. Eur J Haematol 2019; 103:433-441. [PMID: 31381187 DOI: 10.1111/ejh.13308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/25/2019] [Accepted: 07/27/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVES This study aimed to identify the natural course of cytomegalovirus (CMV)/Epstein-Barr virus (EBV) after rabbit antithymocyte globulin and cyclosporine (rATG-CsA) for aplastic anemia (AA). METHODS In 113 prospectively observed AA patients treated with rATG-CsA, the CMV/EBV cohort was classified into two groups by baseline viremic status: no viremia (CMV-G1, n = 112; EBV-G1, n = 98) and the presence of viremia (CMV-G2, n = 1; EBV-G2, n = 13). RESULTS In CMV-G1, the mean CMV load increased up to 3 months but was completely resolved from 6 months. The mean EBV load of EBV-G1 showed a peak at 1 month and then gradually decreased over time but remained detectable throughout the observation period. EBV-G2 showed fluctuating EBV dynamics. With reactivation rates of 38.4% in CMV-G1 and 62.2% in EBV-G1, a longer time to rATG-CsA from diagnosis and a lower absolute lymphocyte count at 1 month from rATG-CsA were significantly associated with CMV and EBV reactivation, respectively. The mean peak CMV and EBV loads of patients with CMV-related (3.5%) and EBV-related (0.9%) diseases were evidently higher than those of the remaining patients without CMV and EBV diseases in the respective cohort. CONCLUSION Considering frequent reactivation and distinct courses of CMV/EBV, virologic surveillance is recommended after rATG-CsA for AA.
Collapse
Affiliation(s)
- Sung-Soo Park
- Division of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Yeon Cho
- Division of Infectious Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eunhee Han
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Gi June Min
- Division of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Silvia Park
- Division of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae-Ho Yoon
- Division of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Eun Lee
- Division of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Byung-Sik Cho
- Division of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ki-Seong Eom
- Division of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yoo-Jin Kim
- Division of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seok Lee
- Division of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hee-Je Kim
- Division of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chang-Ki Min
- Division of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seok-Goo Cho
- Division of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jong Wook Lee
- Division of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
12
|
Kerr JR. Epstein-Barr virus (EBV) reactivation and therapeutic inhibitors. J Clin Pathol 2019; 72:651-658. [DOI: 10.1136/jclinpath-2019-205822] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 05/19/2019] [Accepted: 06/10/2019] [Indexed: 12/11/2022]
Abstract
Epstein-Barr virus (EBV) is a ubiquitous human virus which infects almost all humans during their lifetime and following the acute phase, persists for the remainder of the life of the individual. EBV infects B lymphocytes leading to their immortalisation, with persistence of the EBV genome as an episome. In the latent phase, EBV is prevented from reactivating through efficient cytotoxic cellular immunity. EBV reactivates (lytic phase) under conditions of psychological stress with consequent weakening of cellular immunity, and EBV reactivation has been shown to occur in a subset of individuals with each of a variety of cancers, autoimmune diseases, the autoimmune-like disease, chronic fatigue syndrome/myalgic encephalitis and under other circumstances such as being an inpatient in an intensive care unit. Chronic EBV reactivation is an important mechanism in the pathogenesis of many such diseases, yet is rarely tested for in immunocompetent individuals. This review summarises the pathogenesis of EBV infection, EBV reactivation and its role in disease, and methods which may be used to detect it. Known inhibitors of EBV reactivation and replication are discussed, including drugs licensed for treatment of other herpesviruses, licensed or experimental drugs for various other indications, compounds at an early stage of drug development and nutritional constituents such as vitamins and dietary supplements.
Collapse
|
13
|
Risk Factors and Predictive Scoring System For Post-Transplant Lymphoproliferative Disorder after Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2019; 25:1441-1449. [DOI: 10.1016/j.bbmt.2019.02.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/12/2019] [Indexed: 11/24/2022]
|
14
|
Guan Y, Jakimovski D, Ramanathan M, Weinstock-Guttman B, Zivadinov R. The role of Epstein-Barr virus in multiple sclerosis: from molecular pathophysiology to in vivo imaging. Neural Regen Res 2019; 14:373-386. [PMID: 30539801 PMCID: PMC6334604 DOI: 10.4103/1673-5374.245462] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 08/31/2018] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is a disease of the central nervous system characterized by inflammation, demyelination, and neuronal damage. Environmental and genetic factors are associated with the risk of developing MS, but the exact cause still remains unidentified. Epstein-Barr virus (EBV), vitamin D, and smoking are among the most well-established environmental risk factors in MS. Infectious mononucleosis, which is caused by delayed primary EBV infection, increases the risk of developing MS. EBV may also contribute to MS pathogenesis indirectly by activating silent human endogenous retrovirus-W. The emerging B-cell depleting therapies, particularly anti-CD20 agents such as rituximab, ocrelizumab, as well as the fully human ofatumumab, have shown promising clinical and magnetic resonance imaging benefit. One potential effect of these therapies is the depletion of memory B-cells, the primary reservoir site where EBV latency occurs. In addition, EBV potentially interacts with both genetic and other environmental factors to increase susceptibility and disease severity of MS. This review examines the role of EBV in MS pathophysiology and summarizes the recent clinical and radiological findings, with a focus on B-cells and in vivo imaging. Addressing the potential link between EBV and MS allows the better understanding of MS pathogenesis and helps to identify additional disease biomarkers that may be responsive to B-cell depleting intervention.
Collapse
Affiliation(s)
- Yi Guan
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Murali Ramanathan
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, NY, USA
| | - Bianca Weinstock-Guttman
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
- Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
15
|
Pender MP, Csurhes PA, Smith C, Douglas NL, Neller MA, Matthews KK, Beagley L, Rehan S, Crooks P, Hopkins TJ, Blum S, Green KA, Ioannides ZA, Swayne A, Aftab BT, Hooper KD, Burrows SR, Thompson KM, Coulthard A, Khanna R. Epstein-Barr virus-specific T cell therapy for progressive multiple sclerosis. JCI Insight 2018; 3:124714. [PMID: 30429369 DOI: 10.1172/jci.insight.124714] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/16/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Increasing evidence indicates a role for EBV in the pathogenesis of multiple sclerosis (MS). EBV-infected autoreactive B cells might accumulate in the CNS because of defective cytotoxic CD8+ T cell immunity. We sought to determine the feasibility and safety of treating progressive MS patients with autologous EBV-specific T cell therapy. METHODS An open-label phase I trial was designed to treat 5 patients with secondary progressive MS and 5 patients with primary progressive MS with 4 escalating doses of in vitro-expanded autologous EBV-specific T cells targeting EBV nuclear antigen 1, latent membrane protein 1 (LMP1), and LMP2A. Following adoptive immunotherapy, we monitored the patients for safety and clinical responses. RESULTS Of the 13 recruited participants, 10 received the full course of T cell therapy. There were no serious adverse events. Seven patients showed improvement, with 6 experiencing both symptomatic and objective neurological improvement, together with a reduction in fatigue, improved quality of life, and, in 3 patients, reduced intrathecal IgG production. All 6 patients receiving T cells with strong EBV reactivity showed clinical improvement, whereas only 1 of the 4 patients receiving T cells with weak EBV reactivity showed improvement (P = 0.033, Fisher's exact test). CONCLUSION EBV-specific adoptive T cell therapy was well tolerated. Clinical improvement following treatment was associated with the potency of EBV-specific reactivity of the administered T cells. Further clinical trials are warranted to determine the efficacy of EBV-specific T cell therapy in MS. TRIAL REGISTRATION Australian New Zealand Clinical Trials Registry, ACTRN12615000422527. FUNDING MS Queensland, MS Research Australia, Perpetual Trustee Company Ltd., and donations from private individuals who wish to remain anonymous.
Collapse
Affiliation(s)
- Michael P Pender
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Peter A Csurhes
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,The University of Queensland Centre for Clinical Research, Brisbane, Queensland, Australia
| | - Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Nanette L Douglas
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Michelle A Neller
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Katherine K Matthews
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Leone Beagley
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Sweera Rehan
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Pauline Crooks
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Tracey J Hopkins
- Internal Medicine Day Treatment Unit, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Stefan Blum
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Kerryn A Green
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Zara A Ioannides
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Andrew Swayne
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Blake T Aftab
- Preclinical Science and Translational Medicine, Atara Biotherapeutics, South San Francisco, California, USA
| | - Kaye D Hooper
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Scott R Burrows
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kate M Thompson
- Department of Psychology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,Faculty of Health and Behavioural Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Alan Coulthard
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Medical Imaging, Royal Brisbane and Women's Hospital, Brisbane,Queensland, Australia
| | - Rajiv Khanna
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
16
|
Zhao L, He R, Long H, Guo B, Jia Q, Qin D, Liu SQ, Wang Z, Xiang T, Zhang J, Tan Y, Huang J, Chen J, Wang F, Xiao M, Gao J, Yang X, Zeng H, Wang X, Hu C, Alexander PB, Symonds ALJ, Yu J, Wan Y, Li QJ, Ye L, Zhu B. Late-stage tumors induce anemia and immunosuppressive extramedullary erythroid progenitor cells. Nat Med 2018; 24:1536-1544. [PMID: 30297899 PMCID: PMC6211844 DOI: 10.1038/s41591-018-0205-5] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/27/2018] [Indexed: 01/13/2023]
Abstract
Impaired immunity in patients with late-stage cancer is not limited to antitumor responses, as demonstrated by poor vaccination protection and high susceptibility to infection1-3. This has been largely attributed to chemotherapy-induced impairment of innate immunity, such as neutropenia2, whereas systemic effects of tumors on hematopoiesis and adoptive immunity remain incompletely understood. Here we observed anemia associated with severe deficiency of CD8+ T cell responses against pathogens in treatment-naive mice bearing large tumors. Specifically, we identify CD45+ erythroid progenitor cells (CD71+TER119+; EPCs) as robust immunosuppressors. CD45+ EPCs, induced by tumor growth-associated extramedullary hematopoiesis, accumulate in the spleen to become a major population, outnumbering regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). The CD45+ EPC transcriptome closely resembles that of MDSCs, and, like MDSCs, reactive oxygen species production is a major mechanism underlying CD45+ EPC-mediated immunosuppression. Similarly, an immunosuppressive CD45+ EPC population was detected in patients with cancer who have anemia. These findings identify a major population of immunosuppressive cells that likely contributes to the impaired T cell responses commonly observed in patients with advanced cancer.
Collapse
Affiliation(s)
- Lintao Zhao
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Ran He
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Bo Guo
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Qingzhu Jia
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Diyuan Qin
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Si-Qi Liu
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Zhongyu Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Tong Xiang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jue Zhang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yulong Tan
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Jiani Huang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Junying Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Fang Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry & Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Minglu Xiao
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Jianbao Gao
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xinxin Yang
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Hao Zeng
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xinxin Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Chunyan Hu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Peter B Alexander
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Alistair L J Symonds
- Institute of Cell and Molecular Science, Barts and London School of Medicine and Dentistry, University of London, London, UK
| | - Jia Yu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry & Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Yisong Wan
- Departement of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Qi-Jing Li
- Department of Immunology, Duke University Medical Center, Durham, NC, USA.
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University, Chongqing, China.
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China. .,Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
17
|
Smith C, Khanna R. Immune-based therapeutic approaches to virus-associated cancers. Curr Opin Virol 2018; 32:24-29. [PMID: 30241042 DOI: 10.1016/j.coviro.2018.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 01/20/2023]
Abstract
It is estimated that 60-70% of cancers associated with infectious agents are linked to viral infections. Both RNA and DNA viruses that can establish persistent infection exploit various mechanisms including host cell immortalization through genomic instability, chronic inflammation and immune escape, to promote oncogenic transformation of human cells. Expression of selected viral proteins in malignant cells provides a unique opportunity to employ targeted therapies that can disrupt the cellular proliferation and prevent collateral damage caused by standard clinical therapies. While vaccination can be used to prevent infection before malignant transformation, immune-based therapies based on adoptive transfer of T cells and/or antibodies have emerged as powerful tools for the treatment of virus-associated cancers. Here we discuss recent advances and future prospects of immune-based therapies for virus-associated cancers.
Collapse
Affiliation(s)
- Corey Smith
- QIMR Centre for Immunotherapy and Vaccine Development and Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Qld, Australia.
| | - Rajiv Khanna
- QIMR Centre for Immunotherapy and Vaccine Development and Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Qld, Australia.
| |
Collapse
|
18
|
Epstein-Barr virus–specific cytokine-induced killer cells for treatment of Epstein-Barr virus–related malignant lymphoma. Cytotherapy 2018; 20:839-850. [DOI: 10.1016/j.jcyt.2018.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/16/2018] [Accepted: 04/22/2018] [Indexed: 12/16/2022]
|
19
|
Williams M, Ariza ME. EBV Positive Diffuse Large B Cell Lymphoma and Chronic Lymphocytic Leukemia Patients Exhibit Increased Anti-dUTPase Antibodies. Cancers (Basel) 2018; 10:E129. [PMID: 29723986 PMCID: PMC5977102 DOI: 10.3390/cancers10050129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/20/2018] [Accepted: 04/25/2018] [Indexed: 12/22/2022] Open
Abstract
The Epstein-Barr virus (EBV), which is a ubiquitous γ-herpesvirus, establishes a latent infection in more than 90% of the global adult population. EBV-associated malignancies have increased by 14.6% over the last 20 years, and account for approximately 1.5% of all cancers worldwide and 1.8% of all cancer deaths. However, the potential involvement/contribution of lytic proteins to the pathophysiology of EBV-associated cancers is not well understood. We have previously demonstrated that the EBV-deoxyuridine triphosphate nucleotidohydrolase (dUTPase) modulates innate and adaptive immune responses by engaging the Toll-Like Receptor 2 (TLR2), which leads to the modulation of downstream genes involved in oncogenesis, chronic inflammation, and in effector T-cell function. Furthermore, examination of serum samples from diffuse large B-cell lymphoma (DLBCL) and chronic lymphocytic leukemia patients revealed the presence of increased levels of anti-dUTPase antibodies in both cohorts compared to controls with the highest levels (3.67-fold increase) observed in DLBCL female cases and the lowest (2.12-fold increase) in DLBCL males. Using computer-generated algorithms, dUTPase amino acid sequence alignments, and functional studies of BLLF3 mutants, we identified a putative amino acid motif involved with TLR2 interaction. These findings suggest that the EBV-dUTPase: TLR2 interaction is a potential molecular target that could be used for developing novel therapeutics (small molecules/vaccines).
Collapse
Affiliation(s)
- Marshall Williams
- Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210, USA.
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH 43210, USA.
| | - Maria Eugenia Ariza
- Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210, USA.
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH 43210, USA.
| |
Collapse
|
20
|
Laurent A, Klich A, Roy P, Lina B, Kassai B, Bacchetta J, Cochat P. Pediatric renal transplantation: A retrospective single-center study on epidemiology and morbidity due to EBV. Pediatr Transplant 2018; 22:e13151. [PMID: 29430795 DOI: 10.1111/petr.13151] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/30/2017] [Indexed: 11/29/2022]
Abstract
Pediatric R-Tx patients are at high risk of developing EBV primary infection. Although high DNA replication is a risk factor for PTLD, some patients develop PTLD with low viral load. In this retrospective single-center study including all pediatric patients having received R-Tx (2003-2012 period), we aimed to identify risk factors for uncontrolled reactions to EBV (defined as the presence of a viral load >10 000 copies/mL or PTLD). A Cox proportional hazard model was performed. A total of 117 patients underwent R-Tx at a mean age of 9.7 ± 5.3 years, 46 of them being seronegative for EBV at the time of R-Tx. During follow-up, 54 patients displayed positive EBV viral load, 22 of whom presenting with primary infection. An uncontrolled reaction to EBV was observed in 24 patients, whilst 4 patients developed PTLD. Univariate and multivariate analyses suggested the following risk factors for an uncontrolled reaction: age below 5 years, graft from a deceased donor, ≥5 HLA mismatches, EBV-seronegative status at the time of R-Tx, and a secondary post-Tx loss of anti-EBNA. Monitoring anti-EBNA after R-Tx may contribute to the early identification of patients at risk for uncontrolled reaction.
Collapse
Affiliation(s)
- A Laurent
- Hospices Civils de Lyon, Service de Néphrologie, Rhumatologie et Dermatologie pédiatriques, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Lyon, France
| | - A Klich
- Hospices Civils de Lyon, Service de Biostatistique et de Bioinformatique, Lyon, France.,Université de Lyon, Lyon, France.,CNRS, UMR 5558, Laboratoire de Biométrie et Biologie Evolutive, Equipe Biostatistique-Santé, Villeurbanne, France
| | - P Roy
- Hospices Civils de Lyon, Service de Biostatistique et de Bioinformatique, Lyon, France.,Université de Lyon, Lyon, France.,CNRS, UMR 5558, Laboratoire de Biométrie et Biologie Evolutive, Equipe Biostatistique-Santé, Villeurbanne, France
| | - B Lina
- Laboratoire de Virologie, Institut des Agents Infectieux IAI, Hospices Civils de Lyon, Hopital de la Croix Rousse, Lyon, France.,CIRI, Centre International de Recherche en Infectiologie, Virpath, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - B Kassai
- Université de Lyon, Lyon, France.,Centre d'Investigation Clinique de Lyon, 1407 Inserm-Hospices Civils de Lyon, Groupement Hospitalier Est, Hôpital Femme-Mère-Enfant, EPICIME, Bron, France
| | - J Bacchetta
- Hospices Civils de Lyon, Service de Néphrologie, Rhumatologie et Dermatologie pédiatriques, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Lyon, France.,CNRS, UMR 5558, Laboratoire de Biométrie et Biologie Evolutive, Equipe Biostatistique-Santé, Villeurbanne, France.,Laboratoire de Virologie, Institut des Agents Infectieux IAI, Hospices Civils de Lyon, Hopital de la Croix Rousse, Lyon, France
| | - P Cochat
- Hospices Civils de Lyon, Service de Néphrologie, Rhumatologie et Dermatologie pédiatriques, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Lyon, France.,Université de Lyon, Lyon, France
| |
Collapse
|
21
|
|
22
|
Hart J, MacHugh ND, Sheldrake T, Nielsen M, Morrison WI. Identification of immediate early gene products of bovine herpes virus 1 (BHV-1) as dominant antigens recognized by CD8 T cells in immune cattle. J Gen Virol 2017; 98:1843-1854. [PMID: 28671533 DOI: 10.1099/jgv.0.000823] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In common with other herpes viruses, bovine herpes virus 1 (BHV-1) induces strong virus-specific CD8 T-cell responses. However, there is a paucity of information on the antigenic specificity of the responding T-cells. The development of a system to generate virus-specific CD8 T-cell lines from BHV-1-immune cattle, employing Theileria-transformed cell lines for antigen presentation, has enabled us to address this issue. Use of this system allowed the study to screen for CD8 T-cell antigens that are efficiently presented on the surface of virus-infected cells. Screening of a panel of 16 candidate viral gene products with CD8 T-cell lines from 3 BHV-1-immune cattle of defined MHC genotypes identified 4 antigens, including 3 immediate early (IE) gene products (ICP4, ICP22 and Circ) and a tegument protein (UL49). Identification of the MHC restriction specificities revealed that the antigens were presented by two or three class I MHC alleles in each animal. Six CD8 T-cell epitopes were identified in the three IE proteins by screening of synthetic peptides. Use of an algorithm (NetMHCpan) that predicts the peptide-binding characteristics of restricting MHC alleles confirmed and, in some cases refined, the identity of the epitopes. Analyses of the epitope specificity of the CD8 T-cell lines showed that a large component of the response is directed against these IE epitopes. The results indicate that these IE gene products are dominant targets of the CD8 T-cell response in BHV-I-immune cattle and hence are prime-candidate antigens for the generation of a subunit vaccine.
Collapse
Affiliation(s)
- Jane Hart
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Niall D MacHugh
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Tara Sheldrake
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Morten Nielsen
- Department of Bio and Health Informatics, Centre for Biological Science Sequence Analysis, The Technical University, Lyngby, Denmark
| | - W Ivan Morrison
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK.,Biotechnological Research Institute, National University of San Martin, San Martin, Buenos Aires, Argentina
| |
Collapse
|
23
|
Kyaw T, Peter K, Li Y, Tipping P, Toh BH, Bobik A. Cytotoxic lymphocytes and atherosclerosis: significance, mechanisms and therapeutic challenges. Br J Pharmacol 2017; 174:3956-3972. [PMID: 28471481 DOI: 10.1111/bph.13845] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 04/02/2017] [Accepted: 04/24/2017] [Indexed: 02/06/2023] Open
Abstract
Cytotoxic lymphocytes encompass natural killer lymphocytes (cells) and cytotoxic T cells that include CD8+ T cells, natural killer (NK) T cells, γ, δ (γδ)-T cells and human CD4 + CD28- T cells. These cells play critical roles in inflammatory diseases and in controlling cancers and infections. Cytotoxic lymphocytes can be activated via a number of mechanisms that may involve dendritic cells, macrophages, cytokines or surface proteins on stressed cells. Upon activation, they secrete pro-inflammatory cytokines as well as anti-inflammatory cytokines, chemokines and cytotoxins to promote inflammation and the development of atherosclerotic lesions including vulnerable lesions, which are strongly implicated in myocardial infarctions and strokes. Here, we review the mechanisms that activate and regulate cytotoxic lymphocyte activity, including activating and inhibitory receptors, cytokines, chemokine receptors-chemokine systems utilized to home to inflamed lesions and cytotoxins and cytokines through which they affect other cells within lesions. We also examine their roles in human and mouse models of atherosclerosis and the mechanisms by which they exert their pathogenic effects. Finally, we discuss strategies for therapeutically targeting these cells to prevent the development of atherosclerotic lesions and vulnerable plaques and the challenge of developing highly targeted therapies that only minimally affect the body's immune system, avoiding the complications, such as increased susceptibility to infections, which are currently associated with many immunotherapies for autoimmune diseases. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Tin Kyaw
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Immunology, Monash University, Melbourne, Vic, Australia
| | - Yi Li
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Peter Tipping
- Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Ban-Hock Toh
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Alex Bobik
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Immunology, Monash University, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| |
Collapse
|
24
|
Poorebrahim M, Salarian A, Najafi S, Abazari MF, Aleagha MN, Dadras MN, Jazayeri SM, Ataei A, Poortahmasebi V. Regulatory network analysis of Epstein-Barr virus identifies functional modules and hub genes involved in infectious mononucleosis. Arch Virol 2017; 162:1299-1309. [PMID: 28155194 DOI: 10.1007/s00705-017-3242-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 01/24/2017] [Indexed: 12/14/2022]
Abstract
Epstein-Barr virus (EBV) is the most common cause of infectious mononucleosis (IM) and establishes lifetime infection associated with a variety of cancers and autoimmune diseases. The aim of this study was to develop an integrative gene regulatory network (GRN) approach and overlying gene expression data to identify the representative subnetworks for IM and EBV latent infection (LI). After identifying differentially expressed genes (DEGs) in both IM and LI gene expression profiles, functional annotations were applied using gene ontology (GO) and BiNGO tools, and construction of GRNs, topological analysis and identification of modules were carried out using several plugins of Cytoscape. In parallel, a human-EBV GRN was generated using the Hu-Vir database for further analyses. Our analysis revealed that the majority of DEGs in both IM and LI were involved in cell-cycle and DNA repair processes. However, these genes showed a significant negative correlation in the IM and LI states. Furthermore, cyclin-dependent kinase 2 (CDK2) - a hub gene with the highest centrality score - appeared to be the key player in cell cycle regulation in IM disease. The most significant functional modules in the IM and LI states were involved in the regulation of the cell cycle and apoptosis, respectively. Human-EBV network analysis revealed several direct targets of EBV proteins during IM disease. Our study provides an important first report on the response to IM/LI EBV infection in humans. An important aspect of our data was the upregulation of genes associated with cell cycle progression and proliferation.
Collapse
Affiliation(s)
- Mansour Poorebrahim
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Salarian
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeideh Najafi
- Department of Microbiology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Foad Abazari
- Department of Genetics, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Maryam Nouri Aleagha
- Department of Genetics, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Mohammad Nasr Dadras
- Center for Disease Control, Ministry of Health and Medical Education (MOHME), Tehran, Iran
| | - Seyed Mohammad Jazayeri
- Hepatitis B Molecular Laboratory, Department of Virology, School of Public Health, Tehran University of Medical Sciences, PO Box 15155-6446, Tehran, Iran
| | - Atousa Ataei
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Vahdat Poortahmasebi
- Hepatitis B Molecular Laboratory, Department of Virology, School of Public Health, Tehran University of Medical Sciences, PO Box 15155-6446, Tehran, Iran.
| |
Collapse
|
25
|
Pender MP, Csurhes PA, Burrows JM, Burrows SR. Defective T-cell control of Epstein-Barr virus infection in multiple sclerosis. Clin Transl Immunology 2017; 6:e126. [PMID: 28197337 PMCID: PMC5292561 DOI: 10.1038/cti.2016.87] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/02/2016] [Accepted: 12/02/2016] [Indexed: 02/07/2023] Open
Abstract
Mounting evidence indicates that infection with Epstein–Barr virus (EBV) has a
major role in the pathogenesis of multiple sclerosis (MS). Defective elimination of
EBV-infected B cells by CD8+ T cells might cause MS by allowing
EBV-infected autoreactive B cells to accumulate in the brain. Here we undertake a
comprehensive analysis of the T-cell response to EBV in MS, using flow cytometry and
intracellular IFN-γ staining to measure T-cell responses to EBV-infected
autologous lymphoblastoid cell lines and pools of human leukocyte antigen
(HLA)-class-I-restricted peptides from EBV lytic or latent proteins and
cytomegalovirus (CMV), in 95 patients and 56 EBV-seropositive healthy subjects. In 20
HLA-A2+ healthy subjects and 20 HLA-A2+ patients
we also analysed CD8+ T cells specific for individual peptides,
measured by binding to HLA-peptide complexes and production of IFN-γ,
TNF-α and IL-2. We found a decreased CD8+ T-cell response to
EBV lytic, but not CMV lytic, antigens at the onset of MS and at all subsequent
disease stages. CD8+ T cells directed against EBV latent antigens
were increased but had reduced cytokine polyfunctionality indicating T-cell
exhaustion. During attacks the EBV-specific CD4+ and
CD8+ T-cell populations expanded, with increased functionality of
latent-specific CD8+ T cells. With increasing disease duration,
EBV-specific CD4+ and CD8+ T cells progressively
declined, consistent with T-cell exhaustion. The anti-EBNA1 IgG titre correlated
inversely with the EBV-specific CD8+ T-cell frequency. We postulate
that defective CD8+ T-cell control of EBV reactivation leads to an
expanded population of latently infected cells, including autoreactive B cells.
Collapse
Affiliation(s)
- Michael P Pender
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Cellular ImmunoIogy Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Peter A Csurhes
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Cellular ImmunoIogy Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; The University of Queensland Centre for Clinical Research, Brisbane, Queensland, Australia
| | - Jacqueline M Burrows
- Cellular ImmunoIogy Laboratory, QIMR Berghofer Medical Research Institute , Brisbane, Queensland, Australia
| | - Scott R Burrows
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Cellular ImmunoIogy Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
26
|
Williams MV, Cox B, Ariza ME. Herpesviruses dUTPases: A New Family of Pathogen-Associated Molecular Pattern (PAMP) Proteins with Implications for Human Disease. Pathogens 2016; 6:pathogens6010002. [PMID: 28036046 PMCID: PMC5371890 DOI: 10.3390/pathogens6010002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 12/16/2016] [Accepted: 12/21/2016] [Indexed: 12/13/2022] Open
Abstract
The human herpesviruses are ubiquitous viruses and have a prevalence of over 90% in the adult population. Following a primary infection they establish latency and can be reactivated over a person's lifetime. While it is well accepted that human herpesviruses are implicated in numerous diseases ranging from dermatological and autoimmune disease to cancer, the role of lytic proteins in the pathophysiology of herpesvirus-associated diseases remains largely understudies. Only recently have we begun to appreciate the importance of lytic proteins produced during reactivation of the virus, in particular the deoxyuridine triphosphate nucleotidohydrolases (dUTPase), as key modulators of the host innate and adaptive immune responses. In this review, we provide evidence from animal and human studies of the Epstein-Barr virus as a prototype, supporting the notion that herpesviruses dUTPases are a family of proteins with unique immunoregulatory functions that can alter the inflammatory microenvironment and thus exacerbate the immune pathology of herpesvirus-related diseases including myalgic encephalomyelitis/chronic fatigue syndrome, autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Marshall V Williams
- Department of Cancer Biology and Genetics, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA.
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH 43210, USA.
| | - Brandon Cox
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH 43210, USA.
| | - Maria Eugenia Ariza
- Department of Cancer Biology and Genetics, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA.
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
27
|
Jones AP, Kermode AG, Lucas RM, Carroll WM, Nolan D, Hart PH. Circulating immune cells in multiple sclerosis. Clin Exp Immunol 2016; 187:193-203. [PMID: 27689339 DOI: 10.1111/cei.12878] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2016] [Indexed: 12/17/2022] Open
Abstract
Circulating T and B lymphocytes contribute to the pathogenesis of the neuroinflammatory autoimmune disease, multiple sclerosis (MS). Further progress in the development of MS treatments is dependent upon a greater understanding of the immunological disturbances that underlie the disease. Analyses of circulating immune cells by flow cytometry have revealed MS-associated alterations in the composition and function of T and B cell subsets, including temporal changes associated with disease activity. Disturbances in circulating immune populations reflect those observed in the central nervous system and include skewing towards proinflammatory CD4+ and CD8+ T cells and B cells, greater proportions of follicular T helper cells and functional defects in the corresponding T and B regulatory subsets. Utilizing the analytical power of modern flow cytometers, researchers are now well positioned to monitor immunological changes associated with disease activity or intervention, describe immunological signatures with predictive value and identify targets for therapeutic drug development. This review discusses the contribution of various T and B lymphocyte subsets to MS pathogenesis, provides current and relevant phenotypical descriptions to assist in experimental design and highlights areas of future research.
Collapse
Affiliation(s)
- A P Jones
- Telethon Kids Institute, The University of Western Australia, Perth, WA
| | - A G Kermode
- Centre for Neuromuscular and Neurological Disorders, Western Australian Neuroscience Research Institute, The University of Western Australia, Sir Charles Gairdner Hospital, Perth, WA.,Institute for Immunology and Infectious Disease, Murdoch University, Perth, WA
| | - R M Lucas
- National Centre for Epidemiology and Population Health, Research School of Population Health, Australian National University, Canberra, ACT
| | - W M Carroll
- Centre for Neuromuscular and Neurological Disorders, Western Australian Neuroscience Research Institute, The University of Western Australia, Sir Charles Gairdner Hospital, Perth, WA.,Institute for Immunology and Infectious Disease, Murdoch University, Perth, WA
| | - D Nolan
- Institute for Immunology and Infectious Disease, Murdoch University, Perth, WA.,Immunology Department, Royal Perth Hospital, Perth, WA, Australia
| | - P H Hart
- Telethon Kids Institute, The University of Western Australia, Perth, WA
| |
Collapse
|
28
|
Visco C, Falisi E, Young KH, Pascarella M, Perbellini O, Carli G, Novella E, Rossi D, Giaretta I, Cavallini C, Scupoli MT, De Rossi A, D'Amore ESG, Rassu M, Gaidano G, Pizzolo G, Ambrosetti A, Rodeghiero F. Epstein-Barr virus DNA load in chronic lymphocytic leukemia is an independent predictor of clinical course and survival. Oncotarget 2016; 6:18653-63. [PMID: 26087198 PMCID: PMC4621917 DOI: 10.18632/oncotarget.4418] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 05/09/2015] [Indexed: 11/25/2022] Open
Abstract
The relation between Epstein-Barr virus (EBV) DNA load and clinical course of patients with chronic lymphocytic leukemia (CLL) is unknown. We assessed EBV DNA load by quantitative PCR at CLL presentation in mononuclear cells (MNC) of 220 prospective patients that were enrolled and followed-up in two major Institutions. In 20 patients EBV DNA load was also assessed on plasma samples. Forty-one age-matched healthy subjects were tested for EBV DNA load on MNC. Findings were validated in an independent retrospective cohort of 112 patients with CLL. EBV DNA load was detectable in 59%, and high (≥2000 copies/µg DNA) in 19% of patients, but it was negative in plasma samples. EBV DNA load was significantly higher in CLL patients than in healthy subjects (P < .0001). No relation was found between high EBV load and clinical stage or biological variables, except for 11q deletion (P = .004), CD38 expression (P = .003), and NOTCH1 mutations (P = .05). High EBV load led to a 3.14-fold increase in the hazard ratio of death and to a shorter overall survival (OS; P = .001). Poor OS was attributable, at least in part, to shorter time-to-first-treatment (P = .0008), with no higher risk of Richter's transformation or second cancer. Multivariate analysis selected high levels of EBV load as independent predictor of OS after controlling for confounding clinical and biological variables. EBV DNA load at presentation is an independent predictor of OS in patients with CLL.
Collapse
Affiliation(s)
- Carlo Visco
- Department of Cell Therapy and Hematology, San Bortolo Hospital, Vicenza, Italy
| | - Erika Falisi
- Department of Cell Therapy and Hematology, San Bortolo Hospital, Vicenza, Italy
| | - Ken H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Omar Perbellini
- Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Giuseppe Carli
- Department of Cell Therapy and Hematology, San Bortolo Hospital, Vicenza, Italy
| | - Elisabetta Novella
- Department of Cell Therapy and Hematology, San Bortolo Hospital, Vicenza, Italy
| | - Davide Rossi
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Ilaria Giaretta
- Department of Cell Therapy and Hematology, San Bortolo Hospital, Vicenza, Italy
| | - Chiara Cavallini
- Research Center LURM (University Laboratory of Medical Research), University of Verona, Verona, Italy
| | - Maria Teresa Scupoli
- Research Center LURM (University Laboratory of Medical Research), University of Verona, Verona, Italy
| | - Anita De Rossi
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy.,Istituto Oncologico Veneto(IOV)-IRCCS, Padova, Italy
| | | | - Mario Rassu
- Department of Microbiology, San Bortolo Hospital, Vicenza, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Giovanni Pizzolo
- Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Achille Ambrosetti
- Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | | |
Collapse
|
29
|
Kinch A, Sundström C, Tufveson G, Glimelius I. Association between HLA-A1 and -A2 types and Epstein-Barr virus status of post-transplant lymphoproliferative disorder. Leuk Lymphoma 2016; 57:2351-8. [PMID: 27104753 DOI: 10.3109/10428194.2016.1173209] [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: 01/08/2023]
Abstract
The susceptibility to Epstein-Barr virus (EBV)-related post-transplant lymphoproliferative disorder (PTLD) may be affected by the human leukocyte antigen (HLA) type. We investigated HLA-A and HLA-B allele frequencies, focusing on HLA-A1 and -A2, in a population-based case series of EBV + (n = 60) and EBV- (n = 44) PTLD after solid organ transplantation. The proportion of EBV + PTLD was highest in HLA-A1 homozygotes (100%), lower in carriers of HLA-A1/AX (79%), HLA-A1/A2 (55%), HLA-A2/AX (54%), and lowest in HLA-A2 homozygotes (37%). HLA-A1 type was overrepresented (22% versus 7%, p = 0.05) and HLA-A2 type underrepresented (57% versus 80%, p = 0.01) in patients with EBV + compared with EBV - PTLD. EBV + PTLD in HLA-A1 carriers developed almost exclusively in already EBV-seropositive individuals. EBV status of PTLD was not related to any other HLA-A or HLA-B type. Our findings suggest that HLA-A1 carriers may have an increased risk of EBV + PTLD due to a decreased ability to control the latent EBV infection.
Collapse
Affiliation(s)
- Amelie Kinch
- a Department of Medical Sciences, Section of Infectious Diseases , Uppsala University , Uppsala , Sweden
| | - Christer Sundström
- b Department of Immunology, Genetics and Pathology, Pathology , Uppsala University , Uppsala , Sweden
| | - Gunnar Tufveson
- c Department of Surgical Sciences , Uppsala University , Uppsala , Sweden
| | - Ingrid Glimelius
- d Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology , Uppsala University , Uppsala , Sweden
| |
Collapse
|
30
|
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
|
31
|
Hanaoka N, Murata S, Hosoi H, Shimokado A, Mushino T, Kuriyama K, Hatanaka K, Nishikawa A, Kurimoto M, Sonoki T, Muragaki Y, Nakakuma H. B-Cell-Rich T-Cell Lymphoma Associated with Epstein-Barr Virus-Reactivation and T-Cell Suppression Following Antithymocyte Globulin Therapy in a Patient with Severe Aplastic Anemia. Hematol Rep 2015; 7:5906. [PMID: 26487932 PMCID: PMC4591498 DOI: 10.4081/hr.2015.5906] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/10/2015] [Accepted: 09/02/2015] [Indexed: 11/23/2022] Open
Abstract
B-cell lymphoproliferative disorder (B-LPD) is generally characterized by the proliferation of Epstein-Barr virus (EBV)-infected B lymphocytes. We here report the development of EBV-negative B-LPD associated with EBV-reactivation following antithymocyte globulin (ATG) therapy in a patient with aplastic anemia. The molecular autopsy study showed the sparse EBV-infected clonal T cells could be critically involved in the pathogenesis of EBV-negative oligoclonal B-LPD through cytokine amplification and escape from T-cell surveillances attributable to ATG-based immunosuppressive therapy, leading to an extremely rare B-cell-rich T-cell lymphoma. This report helps in elucidating the complex pathophysiology of intractable B-LPD refractory to rituximab.
Collapse
Affiliation(s)
| | | | | | - Aiko Shimokado
- First Department of Pathology, Wakayama Medical University , Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Epstein-Barr virus latent membrane protein 2A mediated activation of Sonic Hedgehog pathway induces HLA class Ia downregulation in gastric cancer cells. Virology 2015; 484:22-32. [PMID: 26057149 DOI: 10.1016/j.virol.2015.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 05/05/2015] [Accepted: 05/11/2015] [Indexed: 12/12/2022]
Abstract
One of the immune evasion strategies manifested by malignant cells is the downregulation of the Human Leukocyte Antigen (HLA). HLA Class I (HLA- A, -B, -C) present endogenous peptides including viral and tumor antigens to cytotoxic T lymphocytes for immune mediated destruction. We have found the Epstein Barr Virus (EBV) Latent Membrane Protein 2A (LMP2A) to be responsible for this HLA downregulation in gastric cancer cells. Our results further indicate the Sonic Hedgehog pathway; primarily Gli1 to bring about the LMP2A mediated decrease in HLA expression.
Collapse
|
33
|
Smith C, Khanna R. Adoptive therapy for EBV-induced cancers: driving success with post-transplant lymphoproliferative disorder to other EBV-derived tumors. Immunotherapy 2015; 7:563-72. [DOI: 10.2217/imt.15.7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Epstein–Barr virus (EBV) infection is associated with a range of human malignancies of lymphocytic and epithelial cell origin. In addition to viral-mediated and genetic oncogenic events that lead to the establishment of EBV-associated malignancies, defects in the immune control of EBV likely play a significant role in promoting the survival of malignant cells. This breakdown in immune surveillance is most evident in immunocompromised transplant patients who are susceptible to the development of post-transplant lymphoproliferative disorders. Observations over the last two decades have shown that reconstitution of EBV-specific cellular immunity via adoptive cell therapy can have a dramatic effect on both preventing and treating post-transplant lymphoproliferative disorders, leading to hope that similar strategies could be effective in preventing more prevalent EBV-associated malignancies.
Collapse
Affiliation(s)
- Corey Smith
- QIMR Berghofer, Tumour Immunology Laboratory, Department of Immunology, 300 Herston Rd, Brisbane 4006, Australia
| | - Rajiv Khanna
- QIMR Berghofer, Tumour Immunology Laboratory, Department of Immunology, 300 Herston Rd, Brisbane 4006, Australia
| |
Collapse
|
34
|
Abstract
Endemic Burkitt's lymphoma (BL) remains the most prevalent pediatric cancer in sub-Saharan Africa even though it was the first human cancer with a viral etiology described over 50 years ago. Epstein-Barr virus (EBV) was discovered in a BL tumor in 1964 and has since been implicated in other malignancies. The etiology of endemic BL has been linked to EBV and Plasmodium falciparum malaria co-infection. While epidemiologic studies have yielded insight into EBV infection and the etiology of endemic BL, the modulation of viral persistence in children by malaria and deficits in EBV immunosurveillance has more recently been reified. Renewed efforts to design prophylactic and therapeutic EBV vaccines provide hope of preventing EBV-associated BL as well as increasing the ability to cure this cancer.
Collapse
Affiliation(s)
- Rosemary Rochford
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, USA.
| | - Ann M Moormann
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
35
|
Pender MP, Burrows SR. Epstein-Barr virus and multiple sclerosis: potential opportunities for immunotherapy. Clin Transl Immunology 2014; 3:e27. [PMID: 25505955 PMCID: PMC4237030 DOI: 10.1038/cti.2014.25] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 09/30/2014] [Accepted: 09/30/2014] [Indexed: 01/04/2023] Open
Abstract
Multiple sclerosis (MS) is a common chronic inflammatory demyelinating disease of the central nervous system (CNS) causing progressive disability. Many observations implicate Epstein–Barr virus (EBV) in the pathogenesis of MS, namely universal EBV seropositivity, high anti-EBV antibody levels, alterations in EBV-specific CD8+ T-cell immunity, increased spontaneous EBV-induced transformation of peripheral blood B cells, increased shedding of EBV from saliva and accumulation of EBV-infected B cells and plasma cells in the brain. Several mechanisms have been postulated to explain the role of EBV in the development of MS including cross-reactivity between EBV and CNS antigens, bystander damage to the CNS by EBV-specific CD8+ T cells, activation of innate immunity by EBV-encoded small RNA molecules in the CNS, expression of αB-crystallin in EBV-infected B cells leading to a CD4+ T-cell response against oligodendrocyte-derived αB-crystallin and EBV infection of autoreactive B cells, which produce pathogenic autoantibodies and provide costimulatory survival signals to autoreactive T cells in the CNS. The rapidly accumulating evidence for a pathogenic role of EBV in MS provides ground for optimism that it might be possible to prevent and cure MS by effectively controlling EBV infection through vaccination, antiviral drugs or treatment with EBV-specific cytotoxic CD8+ T cells. Adoptive immunotherapy with in vitro-expanded autologous EBV-specific CD8+ T cells directed against viral latent proteins was recently used to treat a patient with secondary progressive MS. Following the therapy, there was clinical improvement, decreased disease activity on magnetic resonance imaging and reduced intrathecal immunoglobulin production.
Collapse
Affiliation(s)
- Michael P Pender
- School of Medicine, The University of Queensland , Brisbane, QLD, Australia ; Department of Neurology, Royal Brisbane and Women's Hospital , Brisbane, QLD, Australia ; QIMR Berghofer Medical Research Institute , Brisbane, QLD, Australia
| | - Scott R Burrows
- School of Medicine, The University of Queensland , Brisbane, QLD, Australia ; QIMR Berghofer Medical Research Institute , Brisbane, QLD, Australia
| |
Collapse
|
36
|
Tellam JT, Zhong J, Lekieffre L, Bhat P, Martinez M, Croft NP, Kaplan W, Tellam RL, Khanna R. mRNA Structural constraints on EBNA1 synthesis impact on in vivo antigen presentation and early priming of CD8+ T cells. PLoS Pathog 2014; 10:e1004423. [PMID: 25299404 PMCID: PMC4192603 DOI: 10.1371/journal.ppat.1004423] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 08/26/2014] [Indexed: 11/18/2022] Open
Abstract
Recent studies have shown that virally encoded mRNA sequences of genome maintenance proteins from herpesviruses contain clusters of unusual structural elements, G-quadruplexes, which modulate viral protein synthesis. Destabilization of these G-quadruplexes can override the inhibitory effect on self-synthesis of these proteins. Here we show that the purine-rich repetitive mRNA sequence of Epstein-Barr virus encoded nuclear antigen 1 (EBNA1) comprising G-quadruplex structures, limits both the presentation of MHC class I-restricted CD8+ T cell epitopes by CD11c+ dendritic cells in draining lymph nodes and early priming of antigen-specific CD8+ T-cells. Destabilization of the G-quadruplex structures through codon-modification significantly enhanced in vivo antigen presentation and activation of virus-specific T cells. Ex vivo imaging of draining lymph nodes by confocal microscopy revealed enhanced antigen-specific T-cell trafficking and APC-CD8+ T-cell interactions in mice primed with viral vectors encoding a codon-modified EBNA1 protein. More importantly, these antigen-specific T cells displayed enhanced expression of the T-box transcription factor and superior polyfunctionality consistent with the qualitative impact of translation efficiency. These results provide an important insight into how viruses exploit mRNA structure to down regulate synthesis of their viral maintenance proteins and delay priming of antigen-specific T cells, thereby establishing a successful latent infection in vivo. Furthermore, targeting EBNA1 mRNA rather than protein by small molecules or antisense oligonucleotides will enhance EBNA1 synthesis and the early priming of effector T cells, to establish a more rapid immune response and prevent persistent infection. Maintenance proteins of viruses establishing latent infections regulate their synthesis to levels sufficient for maintaining persistent infection but below threshold levels for host immune detection. The Epstein-Barr virus maintenance protein, EBNA1, has recently been shown to contain unusual G-quadruplex structures within its repeat mRNA that reduces its translational efficiency. In this study we assess how modification of the EBNA1 mRNA repeat sequence to destabilize the native G-quadruplex structures and thereby increase translation, impacts on the activation of EBNA1-specific T cells in vivo. Mice primed with viral vectors encoding a more efficiently translated EBNA1 mRNA revealed increased trafficking of EBNA1-specific T cells, an enhanced functional profile and increased expression of transcription factors providing evidence for a potential link between mRNA translational efficiency and antigen presentation in vivo and the resultant impact on the functional programming of effector T cells. These findings suggest a novel approach to therapeutic development through the use of antisense strategies or small molecules targeting EBNA1 mRNA structure.
Collapse
Affiliation(s)
- Judy T. Tellam
- QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- * E-mail: (JTT); (RK)
| | - Jie Zhong
- QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Lea Lekieffre
- QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Purnima Bhat
- Medical School, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Michelle Martinez
- QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Nathan P. Croft
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Warren Kaplan
- Peter Wills Bioinformatic Centre, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Ross L. Tellam
- CSIRO Agriculture Flagship, Commonwealth Scientific and Industrial Research Organization, Brisbane, Queensland, Australia
| | - Rajiv Khanna
- QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- * E-mail: (JTT); (RK)
| |
Collapse
|
37
|
Murat P, Tellam J. Effects of messenger RNA structure and other translational control mechanisms on major histocompatibility complex-I mediated antigen presentation. WILEY INTERDISCIPLINARY REVIEWS-RNA 2014; 6:157-71. [PMID: 25264139 PMCID: PMC4359683 DOI: 10.1002/wrna.1262] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 07/18/2014] [Accepted: 07/23/2014] [Indexed: 12/21/2022]
Abstract
Effective T‐cell surveillance of antigen‐presenting cells is dependent on the expression of an array of antigenic peptides bound to major histocompatibility complex (MHC) class I (MHC‐I) or class II (MHC‐II) molecules. Pathogens co‐evolving with their hosts exploit crucial translational regulatory mechanisms in order to evade host immune recognition and thereby sustain their infection. Evasion strategies that downregulate viral protein synthesis and thereby restrict antigen presentation to cytotoxic T‐cells through the endogenous MHC‐I pathway have been implicated in the pathogenesis of viral‐associated malignancies. An understanding of the mechanisms by which messenger RNA (mRNA) structure modulates both viral mRNA translation and the antigen processing machinery to escape immune surveillance, will stimulate the development of alternative therapeutic strategies focused on RNA‐directed drugs designed to enhance immune responses against infected cells. In this review, we discuss regulatory aspects of the MHC‐I pathway and summarize current knowledge of the role attributed by mRNA structure and other translational regulatory mechanisms in immune evasion. In particular we highlight the impact of recently identified G‐quadruplex structures within virally encoded transcripts as unique regulatory signals for translational control and antigen presentation. WIREs RNA 2015, 6:157–171. doi: 10.1002/wrna.1262 This article is categorized under:
RNA Structure and Dynamics > Influence of RNA Structure in Biological Systems Translation > Translation Regulation
Collapse
Affiliation(s)
- Pierre Murat
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
38
|
Chang KC, Chang Y, Wang LHC, Tsai HW, Huang W, Su IJ. Pathogenesis of virus-associated human cancers: Epstein–Barr virus and hepatitis B virus as two examples. J Formos Med Assoc 2014; 113:581-90. [DOI: 10.1016/j.jfma.2013.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 08/02/2013] [Accepted: 09/02/2013] [Indexed: 12/12/2022] Open
|
39
|
Mensen A, Na IK, Häfer R, Meerbach A, Schlecht M, Pietschmann ML, Gruhn B. Comparison of different rabbit ATG preparation effects on early lymphocyte subset recovery after allogeneic HSCT and its association with EBV-mediated PTLD. J Cancer Res Clin Oncol 2014; 140:1971-80. [PMID: 24962343 DOI: 10.1007/s00432-014-1742-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 06/08/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE Rabbit antithymocyte globulin (ATG) is commonly used before allogeneic hematopoietic stem cell transplantation (allo-HSCT) to prevent graft-versus-host disease. Studies comparing the effect of different ATG preparations and dosages on immune reconstitution and risk for Epstein-Barr virus (EBV)-mediated post-transplant lymphoproliferative disorder (PTLD) are rare. METHODS In this retrospective study, we determined T and B cell subsets by flow cytometry after allo-HSCT in children, who received ATG-Genzyme (ATG-G, n = 15), ATG-Fresenius (ATG-F, n = 25) or no-ATG treatment (n = 19). Additionally, PCR-quantified EBV-genome copy counts were correlated with incidence of PTLD. RESULTS We could confirm a dose-dependent impairment of CD8(+) and CD4(+) T cell regeneration by ATG-G, including naïve and memory CD4(+) T cells. No differences were seen between the currently applied dosages of 5-10 mg/kg ATG-G and 20-60 mg/kg ATG-F. Significantly delayed T cell subset reconstitution was determined only at high dosages of 20-60 mg/kg ATG-G compared to ATG-F. B cell reconstitution was comparably impaired in ATG-G- and ATG-F-treated patients. Although the incidence of EBV reactivation was similar in both ATG groups, EBV copy counts of >10(4) copies/10(5) peripheral blood mononuclear cells and the occurrence of PTLD were only found in ATG-G-treated patients. CONCLUSIONS We conclude that high, but importantly not currently applied low dosages of ATG-G, impair thymic T cell regeneration and memory T cell immunity to a greater extent than ATG-F in pediatric patients. In addition, our results suggest an increased risk for EBV-PTLD when treated with ATG-G. Prospective studies are warranted to compare different ATG preparations with regard to the immune reconstitution and EBV-PTLD.
Collapse
Affiliation(s)
- Angela Mensen
- Institute of Medical Immunology, Charité CVK, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Halliday N, Smith C, Atkinson C, O'Beirne J, Patch D, Burroughs AK, Thorburn D, Haque T. Characteristics of Epstein-Barr viraemia in adult liver transplant patients: A retrospective cohort study. Transpl Int 2014; 27:838-46. [DOI: 10.1111/tri.12342] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 11/25/2013] [Accepted: 04/14/2014] [Indexed: 11/27/2022]
Affiliation(s)
- Neil Halliday
- Department of Virology; Royal Free London NHS Foundation Trust; London UK
| | - Colette Smith
- Research Department of Infection and Population Health; UCL; London UK
| | - Claire Atkinson
- Department of Virology; Royal Free London NHS Foundation Trust; London UK
| | - James O'Beirne
- Institute of Liver and Digestive Health; Royal Free London NHS Foundation Trust; London UK
| | - David Patch
- Institute of Liver and Digestive Health; Royal Free London NHS Foundation Trust; London UK
| | - Andrew K Burroughs
- Institute of Liver and Digestive Health; Royal Free London NHS Foundation Trust; London UK
| | - Douglas Thorburn
- Institute of Liver and Digestive Health; Royal Free London NHS Foundation Trust; London UK
| | - Tanzina Haque
- Department of Virology; Royal Free London NHS Foundation Trust; London UK
| |
Collapse
|
41
|
Pender MP, Csurhes PA, Pfluger CM, Burrows SR. Deficiency of CD8+ effector memory T cells is an early and persistent feature of multiple sclerosis. Mult Scler 2014; 20:1825-32. [PMID: 24842963 PMCID: PMC4361480 DOI: 10.1177/1352458514536252] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Patients with multiple sclerosis (MS) have a deficiency of circulating CD8+ T cells, which might impair control of Epstein-Barr virus (EBV) and predispose to MS by allowing EBV-infected autoreactive B cells to accumulate in the central nervous system. Based on the expression of CD45RA and CD62L, CD4+ T cells and CD8+ T cells can be subdivided into four subsets with distinct homing and functional properties, namely: naïve, central memory, effector memory (EM) and effector memory re-expressing CD45RA (EMRA) cells. OBJECTIVE Our aim was to determine which memory subsets are involved in the CD8+ T cell deficiency and how these relate to clinical course. METHODS We used flow cytometry to analyze the memory phenotypes of T cells in the blood of 118 MS patients and 112 healthy subjects. RESULTS MS patients had a decreased frequency of EM (CD45RA(-)CD62L(-)) and EMRA (CD45RA(+)CD62L(-)) CD8+ T cells, which was present at the onset of disease and persisted throughout the clinical course. The frequencies of CD4+ EM and EMRA T cells were normal. CONCLUSION Deficiency of effector memory CD8+ T cells is an early and persistent feature of MS and might underlie the impaired CD8+ T cell control of EBV.
Collapse
Affiliation(s)
- Michael P Pender
- The University of Queensland, School of Medicine, Brisbane, Australia/Royal Brisbane and Women's Hospital, Australia/QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Peter A Csurhes
- The University of Queensland, School of Medicine, Brisbane, Australia/The University of Queensland Centre for Clinical Research, Brisbane, Australia
| | - Casey Mm Pfluger
- The University of Queensland, School of Medicine, Brisbane, Australia/The University of Queensland Centre for Clinical Research, Brisbane, Australia
| | - Scott R Burrows
- The University of Queensland, School of Medicine, Brisbane, Australia/QIMR Berghofer Medical Research Institute, Brisbane, Australia
| |
Collapse
|
42
|
T cells modulate Epstein-Barr virus latency phenotypes during infection of humanized mice. J Virol 2014; 88:3235-45. [PMID: 24390326 DOI: 10.1128/jvi.02885-13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
UNLABELLED Human B cells, the main target of Epstein-Barr virus (EBV), can display several types of latent viral protein expression, denoted 0, I, IIa, IIb, or III. Of these, only type III expression induces proliferation of cells in vitro. These latency types are present at specific stages of infection and are also characteristic of different tumor types, but their generation is not fully understood. In this study, we analyzed the role of T cells in the regulation of EBV viral latency by using humanized NOD/SCID/IL2Rγ(-/-) mice. Several spleens presented macroscopic tumors 4 weeks after infection. Explanted spleen B cells from some of the EBV-infected mice proliferated in vitro, but this was usually lowered when cyclosporine was added to the cultures. This suggested that the in vitro growth of EBV-infected B cells required T cell help; thus, cells other than type III cells were also present in the spleens. Quantitative PCR analysis of promoter activities specific for the different EBV latency types confirmed that in addition to type III cells, type IIa and type I cells were present in the spleen. The relative usage of the viral promoter specific for I and IIa latency types (Q promoter) was higher in CD8(+) cell-depleted mice, and it was absent from CD4(+) cell-depleted mice. These results indicate that CD4(+) T cells are necessary for the generation/maintenance of cells with latency I/IIa in the humanized mice. CD4(+) T cells contributed to this process through their CD40L expression. IMPORTANCE At primary infection with EBV, the infected B cells are proliferating and express viral proteins that have transforming potential. However, when the acute infection is resolved, in healthy individuals EBV is carried by a small fraction of B cells that express a restricted number of viral proteins unable to induce proliferation. Understanding the details of this transition is of fundamental importance. We studied this question in humanized mice by manipulating their different T cell compartments before and during infection with EBV. Our results indicate that CD4(+) T cells are responsible for the switch to a nonproliferating EBV program during primary infection with EBV.
Collapse
|
43
|
Cytotoxic T cell adoptive immunotherapy as a treatment for nasopharyngeal carcinoma. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 21:256-9. [PMID: 24351754 DOI: 10.1128/cvi.00121-13] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Epstein-Barr virus (EBV) is associated with nasopharyngeal carcinoma (NPC). We assess the safety and tolerability of adoptive transfer of autologous cytotoxic T lymphocytes (CTLs) specific for the EBV latent membrane protein (LMP) in a patient with recurrent NPC. After infusion, the majority of pulmonary lesions were no longer evident, although the primary tumor did not regress.
Collapse
|
44
|
De Roos AJ, Martínez-Maza O, Jerome KR, Mirick DK, Kopecky KJ, Madeleine MM, Magpantay L, Edlefsen KL, Lacroix AZ. Investigation of epstein-barr virus as a potential cause of B-cell non-Hodgkin lymphoma in a prospective cohort. Cancer Epidemiol Biomarkers Prev 2013; 22:1747-55. [PMID: 23885038 PMCID: PMC4193346 DOI: 10.1158/1055-9965.epi-13-0240] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND We hypothesized that poor control of Epstein-Barr virus (EBV) infection, leading to reactivation of the virus, increases the risk of non-Hodgkin lymphoma (NHL) in the general population of primarily immunocompetent persons. METHODS We conducted a case-control study nested within the Women's Health Initiative Observational Study cohort in which we measured antibodies to EBV antigens [immunoglobulin G (IgG) to viral capsid antigen (VCA), nuclear antigen (EBNA1), and early antigen-diffuse (EA-D)] and EBV DNA load in prediagnostic samples of 491 B-cell NHL cases and 491 controls. RESULTS We found no association with established EBV infection, based on seropositivity for VCA. Seropositivity for EBNA1 was associated with decreased risk of B-cell NHL, overall [OR = 0.5; 95% confidence interval (CI), 0.3-0.8] and for each of the histologic subtypes examined. Increased risk of chronic lymphocytic leukemia (CLL) and related subtypes was observed with higher levels of EBV DNA and antibody to EA-D, both markers reflective of reactivation. These associations were strongest for cases with the shortest time interval between blood draw and diagnosis. CONCLUSIONS In balance, these results do not provide strong evidence of EBV playing a causal role in B-cell NHL in general population women. The associations we observed may reflect increased risk of NHL with underlying immune impairment or could be due to reverse causation. IMPACT Further characterization of the subtype-specific association with CLL is warranted. Exclusion of cases with preclinical disease markers (such as monoclonal B-lymphocytosis for CLL) may help rule out reverse causation in future studies.
Collapse
Affiliation(s)
- Anneclaire J De Roos
- Authors' Affiliations: Department of Environmental and Occupational Health, Drexel University School of Public Health, Philadelphia, Pennsylvania; Division of Public Health Sciences and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center; Department of Epidemiology, School of Public Health and Department of Laboratory Medicine, University of Washington, Seattle, Washington; Departments of Obstetrics & Gynecology, and Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at UCLA; Jonsson Comprehensive Cancer Center; UCLA AIDS Institute; and Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, California
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Smith C, Khanna R. Immune regulation of human herpesviruses and its implications for human transplantation. Am J Transplant 2013; 13 Suppl 3:9-23; quiz 23. [PMID: 23347211 DOI: 10.1111/ajt.12005] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 07/03/2012] [Accepted: 07/16/2012] [Indexed: 01/25/2023]
Abstract
Human herpesviruses including cytomegalovirus, Epstein-Barr virus, HHV6, HHV7, HHV8, Herpes simplex virus (HSV)-1 and HSV-2 and varicella zoster virus (VZV) have developed an intricate relationship with the human immune system. This is characterized by the interplay between viral immune evasion mechanisms that promote the establishment of a lifelong persistent infection and the induction of a broad humoral and cellular immune response, which prevents the establishment of viral disease. Understanding the immune parameters that control herpesvirus infection, and the strategies the viruses use to evade immune recognition, has been critical in understanding why immunological dysfunction in transplant patients can lead to disease, and in the development of immunological strategies to prevent and control herpesvirus associated diseases.
Collapse
Affiliation(s)
- C Smith
- Australian Centre for Vaccine Development, Tumour Immunology Laboratory, Department of Immunology, Queensland Institute of Medical Research, Brisbane, Australia
| | | |
Collapse
|
46
|
Smith C, Khanna R. Epstein?Barr virus-associated malignancies: pathobiology and emerging therapeutic options. MICROBIOLOGY AUSTRALIA 2013. [DOI: 10.1071/ma13041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
47
|
CD8+ T cells far predominate over CD4+ T cells in healthy immune response to Epstein-Barr virus infected lymphoblastoid cell lines. Blood 2012; 120:5085-7. [DOI: 10.1182/blood-2012-06-437285] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
48
|
Holoendemic malaria exposure is associated with altered Epstein-Barr virus-specific CD8(+) T-cell differentiation. J Virol 2012; 87:1779-88. [PMID: 23175378 DOI: 10.1128/jvi.02158-12] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Coinfection with Plasmodium falciparum malaria and Epstein-Barr virus (EBV) is a major risk factor for endemic Burkitt lymphoma (eBL), still one of the most prevalent pediatric cancers in equatorial Africa. Although malaria infection has been associated with immunosuppression, the precise mechanisms that contribute to EBV-associated lymphomagenesis remain unclear. In this study, we used polychromatic flow cytometry to characterize CD8(+) T-cell subsets specific for EBV-derived lytic (BMFL1 and BRLF1) and latent (LMP1, LMP2, and EBNA3C) antigens in individuals with divergent malaria exposure. No malaria-associated differences in EBV-specific CD8(+) T-cell frequencies were observed. However, based on a multidimensional analysis of CD45RO, CD27, CCR7, CD127, CD57, and PD-1 expression, we found that individuals living in regions with intense and perennial (holoendemic) malaria transmission harbored more differentiated EBV-specific CD8(+) T-cell populations that contained fewer central memory cells than individuals living in regions with little or no (hypoendemic) malaria. This profile shift was most marked for EBV-specific CD8(+) T-cell populations that targeted latent antigens. Importantly, malaria exposure did not skew the phenotypic properties of either cytomegalovirus (CMV)-specific CD8(+) T cells or the global CD8(+) memory T-cell pool. These observations define a malaria-associated aberration localized to the EBV-specific CD8(+) T-cell compartment that illuminates the etiology of eBL.
Collapse
|
49
|
Zhang B, Kracker S, Yasuda T, Casola S, Vanneman M, Hömig-Hölzel C, Wang Z, Derudder E, Li S, Chakraborty T, Cotter SE, Koyama S, Currie T, Freeman GJ, Kutok JL, Rodig SJ, Dranoff G, Rajewsky K. Immune surveillance and therapy of lymphomas driven by Epstein-Barr virus protein LMP1 in a mouse model. Cell 2012; 148:739-51. [PMID: 22341446 DOI: 10.1016/j.cell.2011.12.031] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 09/17/2011] [Accepted: 12/02/2011] [Indexed: 12/21/2022]
Abstract
B cells infected by Epstein-Barr virus (EBV), a transforming virus endemic in humans, are rapidly cleared by the immune system, but some cells harboring the virus persist for life. Under conditions of immunosuppression, EBV can spread from these cells and cause life-threatening pathologies. We have generated mice expressing the transforming EBV latent membrane protein 1 (LMP1), mimicking a constitutively active CD40 coreceptor, specifically in B cells. Like human EBV-infected cells, LMP1+ B cells were efficiently eliminated by T cells, and breaking immune surveillance resulted in rapid, fatal lymphoproliferation and lymphomagenesis. The lymphoma cells expressed ligands for a natural killer (NK) cell receptor, NKG2D, and could be targeted by an NKG2D-Fc fusion protein. These experiments indicate a central role for LMP1 in the surveillance and transformation of EBV-infected B cells in vivo, establish a preclinical model for B cell lymphomagenesis in immunosuppressed patients, and validate a new therapeutic approach.
Collapse
Affiliation(s)
- Baochun Zhang
- Program of Cellular and Molecular Medicine, Children's Hospital and Immune Disease Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Vogl BA, Fagin U, Nerbas L, Schlenke P, Lamprecht P, Jabs WJ. Longitudinal analysis of frequency and reactivity of Epstein-Barr virus-specific T lymphocytes and their association with intermittent viral reactivation. J Med Virol 2012; 84:119-31. [PMID: 22095540 DOI: 10.1002/jmv.22258] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Persistent Epstein-Barr virus (EBV) infection is controlled tightly by virus-specific T cells. EBV infection is reactivated intermittently over time, even in apparently healthy carriers. Changes in frequency and reactivity of memory T cells, particularly of CD8(+) origin, have not been assessed in this context. It is hypothesized that viral reactivation is facilitated by diminished EBV-specific T-cell immunity. To this end, blood samples from 14 healthy donors were collected at irregular time intervals for a period of about 1 year. Samples were screened for both EBV plasma viremia and increases in viral load in PBMCs as parameters of EBV reactivation. PBMCs were subject to IFN-γ ELISPOT analysis using the autologous EBV-transformed lymphoblastoid cell line (EBV-LCL) or appropriate HLA class I-restricted EBV peptides as stimulators. Frequencies of epitope-specific CD8(+) T cells were monitored further using HLA tetramers and flow cytometry. Twelve of 14 donors exhibited signs of asymptomatic EBV reactivation. Viral reactivation was accompanied by either substantially decreased IFN-γ responses against autologous EBV-LCL (eight of 12 study participants) and/or increased responses against particular EBV peptides (six of 12 donors). In seven persons with HLA-A2 and/or -B8 alleles numbers of HLA tetramer-positive CD8(+) T cells also varied over time, but showed no correlation to episodes of detectable viral activity. In summary, IFN-γ reactivity of EBV-specific T cells is not constant. Viral reactivation is detected preferably at times of diminished EBV-LCL-specific cellular immunity. However, increased reactivity of single immunodominant CD8(+) EBV-specific T-cell clones may occur in response to virus replication.
Collapse
Affiliation(s)
- Bastian A Vogl
- Department of Medicine I, University of Luebeck School of Medicine, Luebeck, Germany
| | | | | | | | | | | |
Collapse
|