51
|
Vink P, Delgado Mingorance I, Maximiano Alonso C, Rubio‐Viqueira B, Jung KH, Rodriguez Moreno JF, Grande E, Marrupe Gonzalez D, Lowndes S, Puente J, Kristeleit H, Farrugia D, McNeil SA, Campora L, Di Paolo E, El Idrissi M, Godeaux O, López‐Fauqued M, Salaun B, Heineman TC, Oostvogels L. Immunogenicity and safety of the adjuvanted recombinant zoster vaccine in patients with solid tumors, vaccinated before or during chemotherapy: A randomized trial. Cancer 2019; 125:1301-1312. [PMID: 30707761 PMCID: PMC6766894 DOI: 10.1002/cncr.31909] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/02/2018] [Accepted: 10/18/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND The adjuvanted recombinant zoster vaccine (RZV) has demonstrated >90% efficacy against herpes zoster in adults ≥50 years of age and 68% efficacy in autologous hematopoietic stem cell transplant recipients ≥18 years of age. We report the immunogenicity and safety of RZV administered to patients with solid tumors (STs) before or at the start of a chemotherapy cycle. METHOD In this phase 2/3 observer-blind, multicenter study (NCT01798056), patients with STs who were ≥18 years of age were randomized (1:1) to receive 2 doses of RZV or placebo 1-2 months apart and stratified (4:1) according to the timing of the first dose with respect to the start of a chemotherapy cycle (first vaccination 8-30 days before the start or at the start [±1 day] of a chemotherapy cycle). Anti-glycoprotein E (gE) antibody concentrations, gE-specific CD4+ T cell frequencies, and vaccine response rates (VRRs) were assessed 1 month after dose 1 and 1 and 12 months after dose 2. Reactogenicity and safety were assessed in the total vaccinated cohort through 12 months after dose 2. RESULTS There were 232 participants in the total vaccinated cohort, 185 participants in the according-to-protocol cohort for humoral immunogenicity, and 58 participants in the according-to-protocol cohort for cell-mediated immunogenicity. Postvaccination anti-gE antibody concentrations, gE-specific CD4+ T cell frequencies and VRRs were higher in RZV recipients than in placebo recipients. Solicited adverse events (AEs) were more frequent among RZV recipients than placebo recipients. Incidence of unsolicited AEs, serious AEs, fatalities, and potential immune-mediated diseases were similar between RZV and placebo recipients. CONCLUSION RZV was immunogenic in patients with STs receiving immunosuppressive chemotherapies. Humoral and cell-mediated immune responses persisted 1 year after vaccination. No safety concerns were identified.
Collapse
Affiliation(s)
| | | | | | | | - Kyung Hae Jung
- Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | | | | | | | | | - Javier Puente
- Medical Oncology DepartmentHospital Clínico San CarlosMadridSpain
| | | | - David Farrugia
- Cheltenham General HospitalGloucestershireUnited Kingdom
| | - Shelly A. McNeil
- Canadian Center for Vaccinology, IWK Health Centre and Nova Scotia Health AuthorityDalhousie UniversityHalifaxCanada
| | | | | | | | | | | | | | - Thomas C. Heineman
- GSKKing of PrussiaPennsylvania
- Present address:
Halozyme TherapeuticsSan DiegoCalifornia
| | | | | |
Collapse
|
52
|
van Zyl DG, Mautner J, Delecluse HJ. Progress in EBV Vaccines. Front Oncol 2019; 9:104. [PMID: 30859093 PMCID: PMC6398348 DOI: 10.3389/fonc.2019.00104] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/04/2019] [Indexed: 12/26/2022] Open
Abstract
The Epstein-Barr virus (EBV) is a ubiquitous pathogen that imparts a significant burden of disease on the human population. EBV is the primary cause of infectious mononucleosis and is etiologically linked to the development of numerous malignancies. In recent years, evidence has also been amassed that strongly implicate EBV in the development of several autoimmune diseases, including multiple sclerosis. Prophylactic and therapeutic vaccination has been touted as a possible means of preventing EBV infection and controlling EBV-associated diseases. However, despite several decades of research, no licensed EBV vaccine is available. The majority of EBV vaccination studies over the last two decades have focused on the major envelope protein gp350, culminating in a phase II clinical trial that showed soluble gp350 reduced the incidence of IM, although it was unable to protect against EBV infection. Recently, novel vaccine candidates with increased structural complexity and antigenic content have been developed. The ability of next generation vaccines to safeguard against B-cell and epithelial cell infection, as well as to target infected cells during all phases of infection, is likely to decrease the negative impact of EBV infection on the human population.
Collapse
Affiliation(s)
- Dwain G. van Zyl
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institut National de la Santé et de la Recherche Médicale, Heidelberg, Germany
- German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Josef Mautner
- German Center for Infection Research (DZIF), Heidelberg, Germany
- Children's Hospital, Technische Universität München, and Helmholtz Zentrum München, Bavaria, Germany
| | - Henri-Jacques Delecluse
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institut National de la Santé et de la Recherche Médicale, Heidelberg, Germany
- German Center for Infection Research (DZIF), Heidelberg, Germany
| |
Collapse
|
53
|
Baird NL, Zhu S, Pearce CM, Viejo-Borbolla A. Current In Vitro Models to Study Varicella Zoster Virus Latency and Reactivation. Viruses 2019; 11:v11020103. [PMID: 30691086 PMCID: PMC6409813 DOI: 10.3390/v11020103] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/16/2019] [Accepted: 01/23/2019] [Indexed: 12/26/2022] Open
Abstract
Varicella zoster virus (VZV) is a highly prevalent human pathogen that causes varicella (chicken pox) during primary infection and establishes latency in peripheral neurons. Symptomatic reactivation often presents as zoster (shingles), but it has also been linked to life-threatening diseases such as encephalitis, vasculopathy and meningitis. Zoster may be followed by postherpetic neuralgia, neuropathic pain lasting after resolution of the rash. The mechanisms of varicella zoster virus (VZV) latency and reactivation are not well characterized. This is in part due to the human-specific nature of VZV that precludes the use of most animal and animal-derived neuronal models. Recently, in vitro models of VZV latency and reactivation using human neurons derived from stem cells have been established facilitating an understanding of the mechanisms leading to VZV latency and reactivation. From the models, c-Jun N-terminal kinase (JNK), phosphoinositide 3-kinase (PI3K) and nerve growth factor (NGF) have all been implicated as potential modulators of VZV latency/reactivation. Additionally, it was shown that the vaccine-strain of VZV is impaired for reactivation. These models may also aid in the generation of prophylactic and therapeutic strategies to treat VZV-associated pathologies. This review summarizes and analyzes the current human neuronal models used to study VZV latency and reactivation, and provides some strategies for their improvement.
Collapse
Affiliation(s)
- Nicholas L Baird
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Shuyong Zhu
- Institute of Virology, Hannover Medical School, 30625 Hannover, Germany.
| | - Catherine M Pearce
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | | |
Collapse
|
54
|
Abstract
There are two licensed herpes zoster vaccines. One is a live vaccine (ZVL) based on an attenuated varicella-zoster virus (VZV). The other is a recombinant vaccine (RZV) based on the VZV glycoprotein E (gE) combined with AS01B, a multicomponent adjuvant system. RZV is superior to ZVL in efficacy, and differs from ZVL in that protection is not diminished by the age of the vaccinee and has not waned significantly during 4 years of follow-up. Immunologic studies demonstrated higher peak memory and persistence of T cell responses in RZV compared with ZVL recipients. RZV recipients also showed development and persistence of polyfunctional T cell responses. Taken together, we conclude that the immunologic data parallel and support the higher efficacy over time of RZV compared with ZVL.
Collapse
Affiliation(s)
- Myron J Levin
- a Departments of Pediatrics , University of Colorado Denver School of Medicine, Anschutz Medical Campus , Aurora , CO , USA.,b Department of Medicine , University of Colorado Denver School of Medicine, Anschutz Medical Campus , Aurora , CO , USA
| | - Adriana Weinberg
- a Departments of Pediatrics , University of Colorado Denver School of Medicine, Anschutz Medical Campus , Aurora , CO , USA.,b Department of Medicine , University of Colorado Denver School of Medicine, Anschutz Medical Campus , Aurora , CO , USA.,c Department of Pathology , University of Colorado Denver School of Medicine, Anschutz Medical Campus , Aurora , CO , USA
| |
Collapse
|
55
|
Su HC, Jing H, Angelus P, Freeman AF. Insights into immunity from clinical and basic science studies of DOCK8 immunodeficiency syndrome. Immunol Rev 2019; 287:9-19. [PMID: 30565250 PMCID: PMC6350515 DOI: 10.1111/imr.12723] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 12/29/2022]
Abstract
DOCK8 immunodeficiency syndrome (DIDS) is a progressive combined immunodeficiency that can be distinguished from other combined immunodeficiencies or hyperimmunoglobulinemia E syndromes in featuring (a) profound susceptibility to virus infections of the skin, with associated skin cancers, and (b) severe food allergies. The DOCK8 locus has many repetitive sequence elements that predispose to the generation of large germline deletions as well as recombination-mediated somatic DNA repair. Residual DOCK8 protein contributes to the variable disease phenotype. The severe virus infections of the skin, and probably also VZV-associated vasculopathy, reflect an important function of DOCK8, which is normally required to maintain lymphocyte shape integrity as the cells migrate through dense tissues. Loss of DOCK8 also causes immune deficits through other mechanisms including a milder generalized cell survival defect and skewing of T helper cell subsets. Recent work has uncovered the roles for DOCK8 in dendritic cell responses that can also help explain the virus susceptibility, as well as in regulatory T cells that might help explain autoimmunity in a minority of patients. Fortunately, hematopoietic stem cell transplantation cures the eczema and infection susceptibility of DIDS, but not necessarily the other disease manifestations including food allergies.
Collapse
Affiliation(s)
- Helen C. Su
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health
| | - Huie Jing
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health
| | - Pam Angelus
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health
| |
Collapse
|
56
|
Burke M. Vaccinations for Older Adults. CURRENT GERIATRICS REPORTS 2018. [DOI: 10.1007/s13670-018-0258-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
57
|
Distinctive Roles for Type I and Type II Interferons and Interferon Regulatory Factors in the Host Cell Defense against Varicella-Zoster Virus. J Virol 2018; 92:JVI.01151-18. [PMID: 30089701 DOI: 10.1128/jvi.01151-18] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/02/2018] [Indexed: 02/08/2023] Open
Abstract
Both type I and type II interferons (IFNs) have been implicated in the host defense against varicella-zoster virus (VZV), a common human herpesvirus that causes varicella and zoster. The purpose of this study was to compare their contributions to the control of VZV replication, to identify the signaling pathways that are critical for mediating their antiviral activity, and to define the mechanisms by which the virus counteracts their effects. Gamma interferon (IFN-γ) was much more potent than IFN-α in blocking VZV infection, which was associated with a differential induction of the interferon regulatory factor (IRF) proteins IRF1 and IRF9, respectively. These observations account for the clinical experience that while the formation of VZV skin lesions is initially controlled by local immunity, adaptive virus-specific T cell responses are required to prevent life-threatening VZV infections.IMPORTANCE While both type I and type II IFNs are involved in the control of herpesvirus infections in the human host, to our knowledge, their relative contributions to the restriction of viral replication and spread have not been assessed. We report that IFN-γ has more potent activity than IFN-α against VZV. Findings from this comparative analysis show that the IFN-α-IRF9 axis functions as a first line of defense to delay the onset of viral replication and spread, whereas the IFN-γ-IRF1 axis has the capacity to block the infectious process. Our findings underscore the importance of IRFs in IFN regulation of herpesvirus infection and account for the clinical experience of the initial control of VZV skin infection attributable to IFN-α production, together with the requirement for induction of adaptive IFN-γ-producing VZV-specific T cells to resolve the infection.
Collapse
|
58
|
Sly JR, Harris AL. Recombinant Zoster Vaccine (Shingrix) to Prevent Herpes Zoster. Nurs Womens Health 2018; 22:417-422. [PMID: 30145235 DOI: 10.1016/j.nwh.2018.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/18/2018] [Accepted: 07/01/2018] [Indexed: 06/08/2023]
Abstract
Women ages 50 years and older are at risk for herpes zoster, a reactivated virus from varicella zoster virus (chickenpox) that causes a painful vesicular rash and can result in postherpetic neuralgia. It is estimated that one in three adults will be affected by herpes zoster in their lifetime. Research evidence points to the need to prevent herpes zoster through vaccination. Since 2006, clinicians have been vaccinating adults with zoster vaccine live (brand name Zostavax), but the efficacy of this vaccine wanes with time and advanced age. In October 2017, the U.S. Food and Drug Administration approved recombinant zoster vaccine under the brand name Shingrix to prevent herpes zoster. Studies have shown significantly better efficacy of Shingrix versus Zostavax. This article summarizes new guidance regarding vaccination with Shingrix and discusses implications for women's health.
Collapse
|
59
|
Levin MJ, Kroehl ME, Johnson MJ, Hammes A, Reinhold D, Lang N, Weinberg A. Th1 memory differentiates recombinant from live herpes zoster vaccines. J Clin Invest 2018; 128:4429-4440. [PMID: 30024861 PMCID: PMC6159998 DOI: 10.1172/jci121484] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/13/2018] [Indexed: 12/14/2022] Open
Abstract
The adjuvanted varicella-zoster virus (VZV) glycoprotein E (gE) subunit herpes zoster vaccine (HZ/su) confers higher protection against HZ than the live attenuated zoster vaccine (ZV). To understand the immunologic basis for the different efficacies of the vaccines, we compared immune responses to the vaccines in adults 50 to 85 years old. gE-specific T cells were very low/undetectable before vaccination when analyzed by FluoroSpot and flow cytometry. Both ZV and HZ/su increased gE-specific responses, but at peak memory response (PMR) after vaccination (30 days after ZV or after the second dose of HZ/su), gE-specific CD4+ and CD8+ T cell responses were 10-fold or more higher in HZ/su compared with ZV recipients. Comparing the vaccines, T cell memory responses, including gE-IL-2+ and VZV-IL-2+ spot-forming cells (SFCs), were higher in HZ/su recipients and cytotoxic and effector responses were lower. At 1 year after vaccination, all gE-Th1 and VZV-IL-2+ SFCs remained higher in HZ/su compared with ZV recipients. Mediation analyses showed that IL-2+ PMR were necessary for the persistence of Th1 responses to either vaccine and VZV-IL-2+ PMR explained 73% of the total effect of HZ/su on persistence. This emphasizes the biological importance of the memory responses, which were clearly superior in HZ/su compared with ZV participants.
Collapse
Affiliation(s)
- Myron J. Levin
- Department of Pediatrics, and
- Department of Medicine, University of Colorado School of Medicine
| | - Miranda E. Kroehl
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, and
| | | | - Andrew Hammes
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, and
| | - Dominik Reinhold
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, and
| | | | - Adriana Weinberg
- Department of Pediatrics, and
- Department of Medicine, University of Colorado School of Medicine
- Department of Pathology, University of Colorado School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Denver, Colorado USA
| |
Collapse
|
60
|
Gershon AA. Tale of two vaccines: differences in response to herpes zoster vaccines. J Clin Invest 2018; 128:4245-4247. [PMID: 30179221 DOI: 10.1172/jci123217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
About one-third of the US population will develop herpes zoster (HZ, commonly known as shingles) over a lifetime, while two-thirds will not. It is not clear exactly why certain people are susceptible to HZ; however, we may be coming closer to an answer. In this issue of the JCI, a study by Levin et al. provides important details concerning pathogenesis of and protection from HZ. The authors characterized differences in the immunologic responses induced by two HZ vaccines, the live attenuated zoster vaccine (ZV) and the more recently developed adjuvanted varicella-zoster virus (VZV) glycoprotein E (gE) subunit herpes zoster vaccine (HZ/su), in vaccine-naive subjects and those previously vaccinated with HZ. The observed differences in responses paralleled the observed clinical protection of the two zoster vaccines, with HZ/su being superior to HZ. Together, these results seem to explain immunologically why the new subunit vaccine outperforms the live vaccine. These differences may also provide clues as to why HZ develops in the first place.
Collapse
|
61
|
Como CN, Pearce CM, Cohrs RJ, Baird NL. Interleukin-6 and type 1 interferons inhibit varicella zoster virus replication in human neurons. Virology 2018; 522:13-18. [PMID: 29979960 DOI: 10.1016/j.virol.2018.06.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 06/25/2018] [Accepted: 06/26/2018] [Indexed: 12/18/2022]
Abstract
Varicella zoster virus (VZV) is a neurotropic alphaherpesvirus that, following primary infection (varicella), establishes latency in sensory, autonomic, sympathetic and parasympathetic neurons, where it remains until reactivation (zoster). VZV-specific cell-mediated immune responses maintain VZV latency; thus, immunosuppressed and elderly persons are at risk of reactivation and associated neurological diseases. However, the cytokines produced by the immune system that control VZV in neurons are largely unknown. Therefore, to better understand how the immune system may restrict VZV in neurons, we studied interleukin-6, tumor necrosis factor-alpha and type 1 interferons for their ability to inhibit VZV replication in human neurons in vitro. Our studies revealed that VZV transcription and viral spread were significantly reduced by interleukin-6 and type 1 interferons, and to a lesser extent by tumor necrosis factor-alpha. These findings will help in understanding how the innate immune system limits virus replication in neurons in vivo.
Collapse
Affiliation(s)
- Christina N Como
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Catherine M Pearce
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Randall J Cohrs
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA; Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Nicholas L Baird
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
62
|
Jacqueline C, Bonnefoy N, Charrière GM, Thomas F, Roche B. Personal history of infections and immunotherapy: Unexpected links and possible therapeutic opportunities. Oncoimmunology 2018; 7:e1466019. [PMID: 30221066 PMCID: PMC6136881 DOI: 10.1080/2162402x.2018.1466019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 01/17/2023] Open
Abstract
The recent breakthroughs in the understanding of tumor immune biology have given rise to a new generation of immunotherapies, harnessing the immune system to eliminate tumors. As the typology and frequency of encountered infections are susceptible to shape the immune system, it could also impact the efficiency of immunotherapy. In this review, we report evidences for an indirect link between personal history of infection and different strategies of immunotherapy. In the current context of interest rise for personalized medicine, we discuss the potential medical applications of considering personal history of infection to design immunotherapeutic strategies.
Collapse
Affiliation(s)
- Camille Jacqueline
- Centre for Ecological and Evolutionary Research on Cancer (CREEC), Montpellier, France
- MIVEGEC, IRD, CNRS, Université Montpellier, Montpellier, France
| | - Nathalie Bonnefoy
- IRCM, INSERM, Université de Montpellier, ICM, F-34298, Montpellier, France
| | - Guillaume M. Charrière
- IHPE, UMR 5244, CNRS, Ifremer, Université de Perpignan Via Domitia, Université de Montpellier, Montpellier, 34095, France
| | - Frédéric Thomas
- Centre for Ecological and Evolutionary Research on Cancer (CREEC), Montpellier, France
- MIVEGEC, IRD, CNRS, Université Montpellier, Montpellier, France
| | - Benjamin Roche
- Centre for Ecological and Evolutionary Research on Cancer (CREEC), Montpellier, France
- UMMISCO, IRD/ Sorbonne Université, Bondy, France
- Departamento de Etología, Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| |
Collapse
|
63
|
Martin LK, Hollaus A, Stahuber A, Hübener C, Fraccaroli A, Tischer J, Schub A, Moosmann A. Cross-sectional analysis of CD8 T cell immunity to human herpesvirus 6B. PLoS Pathog 2018; 14:e1006991. [PMID: 29698478 PMCID: PMC5919459 DOI: 10.1371/journal.ppat.1006991] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/26/2018] [Indexed: 12/15/2022] Open
Abstract
Human herpesvirus 6 (HHV-6) is prevalent in healthy persons, causes disease in immunosuppressed carriers, and may be involved in autoimmune disease. Cytotoxic CD8 T cells are probably important for effective control of infection. However, the HHV-6-specific CD8 T cell repertoire is largely uncharacterized. Therefore, we undertook a virus-wide analysis of CD8 T cell responses to HHV-6. We used a simple anchor motif-based algorithm (SAMBA) to identify 299 epitope candidates potentially presented by the HLA class I molecule B*08:01. Candidates were found in 77 of 98 unique HHV-6B proteins. From peptide-expanded T cell lines, we obtained CD8 T cell clones against 20 candidates. We tested whether T cell clones recognized HHV-6-infected cells. This was the case for 16 epitopes derived from 12 proteins from all phases of the viral replication cycle. Epitopes were enriched in certain amino acids flanking the peptide. Ex vivo analysis of eight healthy donors with HLA-peptide multimers showed that the strongest responses were directed against an epitope from IE-2, with a median frequency of 0.09% of CD8 T cells. Reconstitution of T cells specific for this and other HHV-6 epitopes was also observed after allogeneic hematopoietic stem cell transplantation. We conclude that HHV-6 induces CD8 T cell responses against multiple antigens of diverse functional classes. Most antigens against which CD8 T cells can be raised are presented by infected cells. Ex vivo multimer staining can directly identify HHV-6-specific T cells. These results will advance development of immune monitoring, adoptive T cell therapy, and vaccines. This paper deals with the immune response to a very common virus, called human herpesvirus 6 (HHV-6). Most people catch HHV-6 in early childhood, which often leads to a disease known as three-day fever. Later in life, the virus stays in the body, and an active immune response is needed to prevent the virus from multiplying and causing damage. It is suspected that HHV-6 contributes to autoimmune diseases and chronic fatigue. Moreover, patients with severely weakened immune responses, for example after some forms of transplantation, clearly have difficulties controlling HHV-6, which puts them at risk of severe disease and shortens their survival. This can potentially be prevented by giving them HHV-6-specific "killer" CD8 T cells, which are cells of the immune system that destroy body cells harboring the virus. However, little is known so far about such T cells. Here, we describe 16 new structures that CD8 T cells can use to recognize and kill HHV-6-infected cells. We show that very different viral proteins can furnish such structures. We also observe that such T cells are regularly present in healthy people and in transplant patients who control the virus. Our results will help develop therapies of disease due to HHV-6.
Collapse
MESH Headings
- Adult
- Anemia, Aplastic/immunology
- Anemia, Aplastic/therapy
- Antigens, Viral/immunology
- CD8-Positive T-Lymphocytes/immunology
- Case-Control Studies
- Cells, Cultured
- Cross-Sectional Studies
- Epitopes, T-Lymphocyte/immunology
- HLA Antigens/immunology
- Hematopoietic Stem Cell Transplantation
- Herpesvirus 6, Human/immunology
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Roseolovirus Infections/immunology
- Roseolovirus Infections/virology
- T-Lymphocytes, Cytotoxic
- Transplantation, Homologous
Collapse
Affiliation(s)
- Larissa K. Martin
- DZIF Research Group "Host Control of Viral Latency and Reactivation" (HOCOVLAR), Research Unit Gene Vectors, Helmholtz Zentrum München, Munich, Germany
| | - Alexandra Hollaus
- DZIF Research Group "Host Control of Viral Latency and Reactivation" (HOCOVLAR), Research Unit Gene Vectors, Helmholtz Zentrum München, Munich, Germany
| | - Anna Stahuber
- DZIF Research Group "Host Control of Viral Latency and Reactivation" (HOCOVLAR), Research Unit Gene Vectors, Helmholtz Zentrum München, Munich, Germany
| | - Christoph Hübener
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Alessia Fraccaroli
- Internal Medicine III, Hematopoietic Stem Cell Transplantation, Klinikum der Universität München (LMU), Grosshadern, Munich, Germany
| | - Johanna Tischer
- Internal Medicine III, Hematopoietic Stem Cell Transplantation, Klinikum der Universität München (LMU), Grosshadern, Munich, Germany
| | - Andrea Schub
- DZIF Research Group "Host Control of Viral Latency and Reactivation" (HOCOVLAR), Research Unit Gene Vectors, Helmholtz Zentrum München, Munich, Germany
| | - Andreas Moosmann
- DZIF Research Group "Host Control of Viral Latency and Reactivation" (HOCOVLAR), Research Unit Gene Vectors, Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research (DZIF–Deutsches Zentrum für Infektionsforschung), Munich, Germany
- * E-mail:
| |
Collapse
|
64
|
Asian Elephant T Cell Responses to Elephant Endotheliotropic Herpesvirus. J Virol 2018; 92:JVI.01951-17. [PMID: 29263271 PMCID: PMC5827410 DOI: 10.1128/jvi.01951-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 12/14/2017] [Indexed: 12/20/2022] Open
Abstract
Elephant endotheliotropic herpesvirus (EEHV) can cause lethal hemorrhagic disease in juvenile Asian elephants, an endangered species. One hypothesis to explain this vulnerability of some juvenile elephants is that they fail to mount an effective T cell response to the virus. To our knowledge, there have been no studies of Asian elephant T cell responses to EEHV. To address this deficiency, we validated the gamma interferon (IFN-γ) enzyme-linked immunospot assay for tracking antigen-directed T cell activity by monitoring rabies-specific responses in vaccinated elephants. In addition, we generated monoclonal antibodies to Asian elephant CD4 and CD8 to facilitate phenotypic T cell profiling. Using these tools, we screened healthy elephants with a history of EEHV infection for reactivity against nine EEHV proteins whose counterparts in other herpesviruses are known to induce T cell responses in their natural hosts. We identified glycoprotein B (gB) and the putative regulatory protein E40 as the most immunogenic T cell targets (IFN-γ responses in five of seven elephants), followed by the major capsid protein (IFN-γ responses in three of seven elephants). We also observed that IFN-γ responses were largely from CD4+ T cells. We detected no activity against the predicted major immediate early (E44) and large tegument (E34) proteins, both immunodominant T cell targets in humans latently infected with cytomegalovirus. These studies identified EEHV-specific T cells in Asian elephants for the first time, lending insight into the T cell priming that might be required to protect against EEHV disease, and will guide the design of effective vaccine strategies. IMPORTANCE Endangered Asian elephants are facing many threats, including lethal hemorrhagic disease from elephant endotheliotropic herpesvirus (EEHV). EEHV usually establishes chronic, benign infections in mature Asian elephants but can be lethal to juvenile elephants in captivity and the wild. It is the leading cause of death in captive Asian elephants in North America and Europe. Despite the availability of sensitive tests and protocols for treating EEHV-associated illness, these measures are not always effective. The best line of defense would be a preventative vaccine. We interrogated normal healthy elephants previously infected with EEHV for T cell responses to nine EEHV proteins predicted to induce cellular immune responses. Three proteins elicited IFN-γ responses, suggesting their potential usefulness as vaccine candidates. Our work is the first to describe T cell responses to a member of the proposed fourth subfamily of mammalian herpesviruses, the Deltaherpesvirinae, within a host species in the clade Afrotheria. An EEHV vaccine would greatly contribute to the health care of Asian and African elephants that are also susceptible to this disease.
Collapse
|
65
|
Kim CK, Choi YM, Bae E, Jue MS, So HS, Hwang ES. Reduced NK cell IFN-γ secretion and psychological stress are independently associated with herpes zoster. PLoS One 2018; 13:e0193299. [PMID: 29466462 PMCID: PMC5821387 DOI: 10.1371/journal.pone.0193299] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 02/08/2018] [Indexed: 11/17/2022] Open
Abstract
The pathogenesis of herpes zoster is closely linked to reduced varicella-zoster virus-specific cell-mediated immunity. However, little is known about the interplay between natural killer cells and psychological stress in the pathogenesis of herpes zoster. This study aimed to investigate possible associations among natural killer cells, T cells and psychological stress in herpes zoster. Interferon-gamma secretion from natural killer cell, psychological stress events, stress cognition scale scores and cytomegalovirus-specific cell-mediated immunity were compared between 44 patients with herpes zoster and 44 age- and gender-matched control subjects. A significantly lower median level of interferon-gamma secreted by natural killer cells was observed in patients with a recent diagnosis of herpes zoster than in control subjects (582.7 pg/ml vs. 1783 pg/ml; P = 0.004), whereas cytomegalovirus-specific cell-mediated immunity was not associated with herpes zoster. Psychological stress events and high stress cognition scale scores were significantly associated in patients with herpes zoster (P<0.001 and P = 0.037, respectively). However, reduced interferon-gamma secretion from natural killer cell and psychological stress were not associated. In conclusion, patients with a recent diagnosis of herpes zoster display reduced interferon-gamma secretion from natural killer cells and frequent previous psychological stress events compared with controls. However, reduced natural killer cell activity is not an immunological mediator between psychological stress and herpes zoster.
Collapse
Affiliation(s)
- Choon Kwan Kim
- Department of Internal Medicine, Division of Infectious Diseases, Veterans Health Service Medical Center, Seoul, Korea
| | - Youn Mi Choi
- Department of Laboratory Medicine, Veterans Health Service Medical Center, Seoul, Korea
| | - Eunsin Bae
- Department of Laboratory Medicine, Veterans Health Service Medical Center, Seoul, Korea
| | - Mihn Sook Jue
- Department of Dermatology, Veterans Health Service Medical Center, Seoul, Korea
| | - Hyung Seok So
- Department of Psychiatry, Veterans Health Service Medical Center, Seoul, Korea
| | - Eung-Soo Hwang
- Department of Microbiology and Immunology, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, Korea
| |
Collapse
|
66
|
Weyand CM, Berry GJ, Goronzy JJ. The immunoinhibitory PD-1/PD-L1 pathway in inflammatory blood vessel disease. J Leukoc Biol 2017; 103:565-575. [PMID: 28848042 DOI: 10.1189/jlb.3ma0717-283] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/03/2017] [Indexed: 12/16/2022] Open
Abstract
Because of their vital function, the wall structures of medium and large arteries are immunoprivileged and protected from inflammatory attack. That vascular immunoprivilege is broken in atherosclerosis and in vasculitis, when wall-invading T cells and macrophages (Mϕ) promote tissue injury and maladaptive repair. Historically, tissue-residing T cells were studied for their antigen specificity, but recent progress has refocused attention to antigen-nonspecific regulation, which determines tissue access, persistence, and functional differentiation of T cells. The coinhibitory receptor PD-1, expressed on T cells, delivers negative signals when engaged by its ligand PD-L1, expressed on dendritic cells, Mϕ, and endothelial cells to attenuate T cell activation, effector functions, and survival. Through mitigating signals, the PD-1 immune checkpoint maintains tissue tolerance. In line with this concept, dendritic cells and Mϕs from patients with the vasculitic syndrome giant cell arteritis (GCA) are PD-L1lo ; including vessel-wall-embedded DCs that guard the vascular immunoprivilege. GCA infiltrates in the arterial walls are filled with PD-1+ T cells that secrete IFN-γ, IL-17, and IL-21; drive inflammation-associated angiogenesis; and facilitate intimal hyperplasia. Conversely, chronic tissue inflammation in the atherosclerotic plaque is associated with an overreactive PD-1 checkpoint. Plaque-residing Mϕs are PD-L1hi , a defect induced by their addiction to glucose and glycolytic breakdown. PD-L1hi Mϕs render patients with coronary artery disease immunocompromised and suppress antiviral immunity, including protective anti-varicella zoster virus T cells. Thus, immunoinhibitory signals affect several domains of vascular inflammation; failing PD-L1 in vasculitis enables unopposed immunostimulation and opens the flood gates for polyfunctional inflammatory T cells, and excess PD-L1 in the atherosclerotic plaque disables tissue-protective T cell immunity.
Collapse
Affiliation(s)
- Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Gerald J Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Jörg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
67
|
Wei L, Zhao J, Wu W, Zhang Y, Fu X, Chen L, Wang X. Decreased absolute numbers of CD3 + T cells and CD8 + T cells during aging in herpes zoster patients. Sci Rep 2017; 7:15039. [PMID: 29118328 PMCID: PMC5678144 DOI: 10.1038/s41598-017-15390-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/26/2017] [Indexed: 01/27/2023] Open
Abstract
Herpes zoster (HZ) is an infectious dermatosis with high incidence worldwide. Age is a key risk factor for HZ, and postherpetic neuralgia (PHN) is the main sequelae. Until now, no index has been available to predict the pathogenesis of PHN, and rare reports have focused on the immune response during aging and PHN. In this study, we selected immunoglobulin and complement proteins as markers for humoral immunity, while T lymphocyte subsets and natural killer (NK) cells were selected as markers for cell immunity, to systematically study the characteristics of immune responses in the peripheral blood of HZ patients. Our data showed that the absolute number of CD3+ T cells and CD8+ T cells decreased during aging and PHN. This implies that more attention should be paid to prevent the occurrence of PHN, especially in the aged population.
Collapse
Affiliation(s)
- Li Wei
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory of Diagnostic and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianguang Zhao
- Department of Dermatology, The Dermatovenereology Hospital, Quzhou, China
| | - Wei Wu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory of Diagnostic and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Zhang
- Department of Dermatology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuyan Fu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory of Diagnostic and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lifeng Chen
- Department of Dermatology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoting Wang
- Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
68
|
Levin MJ, Bresnitz E, Popmihajlov Z, Weinberg A, Liaw KL, Willis E, Curtis JR. Studies with herpes zoster vaccines in immune compromised patients. Expert Rev Vaccines 2017; 16:1217-1230. [PMID: 29053937 DOI: 10.1080/14760584.2017.1395703] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The active component of the herpes zoster vaccine (ZVL), licensed for people ≥50 years of age, is a live attenuated varicella-zoster virus. ZVL is contraindicated for immune compromised individuals, with limited regard to the degree of immunosuppression. Areas covered: This review evaluates phase I and II and observational studies for ZVL, and published reports of the off-label use of ZVL, for conditions and therapies for which investigators considered the risk-benefit for using ZVL to be favorable. It also discusses exploratory trials of ZVL for additional immune compromising conditions, and summarizes clinical guidelines from many countries and professional societies that are based upon recent investigations. Studies in immune compromised patients of investigational vaccines that do not contain live virus are reviewed. Expert commentary: It is likely that past and ongoing research with ZVL will define immune compromising diseases and/or therapies for which the risk-benefit for using ZVL vaccine is favorable. The main variables to consider in this assessment in immune compromised patients are safety, immunogenicity, protection against herpes zoster, and persistence of protection. Vaccination against herpes zoster prior to suppressing immunity is an important clinical strategy, although efficacy of this approach has not been evaluated in a clinical trial.
Collapse
Affiliation(s)
- Myron J Levin
- a Section of Pediatric Infectious Diseases, Departments of Pediatrics and Medicine , University of Colorado Anschutz Medical Campus , Aurora , CO , USA
| | | | | | - Adriana Weinberg
- a Section of Pediatric Infectious Diseases, Departments of Pediatrics and Medicine , University of Colorado Anschutz Medical Campus , Aurora , CO , USA
| | | | | | - Jeffrey R Curtis
- c Division of Clinical Immunology and Rheumatology , University of Alabama , Birmingham , AL , USA
| |
Collapse
|
69
|
Targeted Genome Sequencing Reveals Varicella-Zoster Virus Open Reading Frame 12 Deletion. J Virol 2017; 91:JVI.01141-17. [PMID: 28747504 DOI: 10.1128/jvi.01141-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 07/21/2017] [Indexed: 12/19/2022] Open
Abstract
The neurotropic herpesvirus varicella-zoster virus (VZV) establishes a lifelong latent infection in humans following primary infection. The low abundance of VZV nucleic acids in human neurons has hindered an understanding of the mechanisms that regulate viral gene transcription during latency. To overcome this critical barrier, we optimized a targeted capture protocol to enrich VZV DNA and cDNA prior to whole-genome/transcriptome sequence analysis. Since the VZV genome is remarkably stable, it was surprising to detect that VZV32, a VZV laboratory strain with no discernible growth defect in tissue culture, contained a 2,158-bp deletion in open reading frame (ORF) 12. Consequently, ORF 12 and 13 protein expression was abolished and Akt phosphorylation was inhibited. The discovery of the ORF 12 deletion, revealed through targeted genome sequencing analysis, points to the need to authenticate the VZV genome when the virus is propagated in tissue culture.IMPORTANCE Viruses isolated from clinical samples often undergo genetic modifications when cultured in the laboratory. Historically, VZV is among the most genetically stable herpesviruses, a notion supported by more than 60 complete genome sequences from multiple isolates and following multiple in vitro passages. However, application of enrichment protocols to targeted genome sequencing revealed the unexpected deletion of a significant portion of VZV ORF 12 following propagation in cultured human fibroblast cells. While the enrichment protocol did not introduce bias in either the virus genome or transcriptome, the findings indicate the need for authentication of VZV by sequencing when the virus is propagated in tissue culture.
Collapse
|
70
|
Perciani CT, Jaoko W, Walmsley S, Farah B, Mahmud SM, Ostrowski M, Anzala O, Team KI, MacDonald KS. Protocol of a randomised controlled trial characterising the immune responses induced by varicella-zoster virus (VZV) vaccination in healthy Kenyan women: setting the stage for a potential VZV-based HIV vaccine. BMJ Open 2017; 7:e017391. [PMID: 28939581 PMCID: PMC5623463 DOI: 10.1136/bmjopen-2017-017391] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION A protective HIV vaccine would be expected to induce durable effector immune responses at the mucosa, restricting HIV infection at its portal of entry. We hypothesise that use of varicella-zoster virus (VZV) as an HIV delivery vector could generate sustained and robust tissue-based immunity against HIV antigens to provide long-term protection against HIV. Given that HIV uniquely targets immune-activated T cells, the development of human vaccines against HIV must also involve a specific examination of the safety of the vector. Thus, we aim to evaluate the effects of VZV vaccination on the recipients' immune activation state, and on VZV-specific circulating humoral and cellular responses in addition to those at the cervical and rectal mucosa. METHODS AND ANALYSIS This open-label, randomised, longitudinal crossover study includes healthy Kenyan VZV-seropositive women at low risk for HIV infection. Participants receive a single dose of a commercial live-attenuated VZVOka vaccine at either week 0 (n=22) or at week 12 (n=22) of the study and are followed for 48 and 36 weeks postvaccination, respectively. The primary outcome is the change on cervical CD4+ T-cell immune activation measured by the coexpression of CD38 and HLA-DR 12 weeks postvaccination compared with the baseline (prevaccination). Secondary analyses include postvaccination changes in VZV-specific mucosal and systemic humoral and cellular immune responses, changes in cytokine and chemokine measures, study acceptability and feasibility of mucosal sampling and a longitudinal assessment of the bacterial community composition of the mucosa. ETHICS AND DISSEMINATION The study has ethical approval from Kenyatta National Hospital/University of Nairobi Ethics and Research Committee, the University of Toronto Research Ethics Board and by Kenyan Pharmacy and Poisons Board. Results will be presented at conferences, disseminated to participants and stakeholders as well as published in peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT02514018. Pre-results.
Collapse
Affiliation(s)
- Catia T Perciani
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Walter Jaoko
- Kenyan AIDS Vaccine Initiative-Institute of Clinical Research (KAVI-ICR), Nairobi, Kenya
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Sharon Walmsley
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Bashir Farah
- Kenyan AIDS Vaccine Initiative-Institute of Clinical Research (KAVI-ICR), Nairobi, Kenya
| | - Salaheddin M Mahmud
- Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Mario Ostrowski
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Omu Anzala
- Kenyan AIDS Vaccine Initiative-Institute of Clinical Research (KAVI-ICR), Nairobi, Kenya
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Kavi-Icr Team
- Kenyan AIDS Vaccine Initiative-Institute of Clinical Research (KAVI-ICR), Nairobi, Kenya
| | - Kelly S MacDonald
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Section of Infectious Diseases, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
71
|
Abstract
The most common specimens from immunocompromised patients that are analyzed for detection of herpes simplex virus (HSV) or varicella-zoster virus (VZV) are from skin lesions. Many types of assays are applicable to these samples, but some, such as virus isolation and direct fluorescent antibody testing, are useful only in the early phases of the lesions. In contrast, nucleic acid (NA) detection methods, which generally have superior sensitivity and specificity, can be applied to skin lesions at any stage of progression. NA methods are also the best choice, and sometimes the only choice, for detecting HSV or VZV in blood, cerebrospinal fluid, aqueous or vitreous humor, and from mucosal surfaces. NA methods provide the best performance when reliability and speed (within 24 hours) are considered together. They readily distinguish the type of HSV detected or the source of VZV detected (wild type or vaccine strain). Nucleic acid detection methods are constantly being improved with respect to speed and ease of performance. Broader applications are under study, such as the use of quantitative results of viral load for prognosis and to assess the efficacy of antiviral therapy.
Collapse
|
72
|
Sahoo F, Hill JA, Xie H, Leisenring W, Yi J, Goyal S, Kimball LE, Lee I, Seo S, Davis C, Pergam SA, Flowers ME, Liaw KL, Holmberg L, Boeckh M. Herpes Zoster in Autologous Hematopoietic Cell Transplant Recipients in the Era of Acyclovir or Valacyclovir Prophylaxis and Novel Treatment and Maintenance Therapies. Biol Blood Marrow Transplant 2016; 23:505-511. [PMID: 28039754 DOI: 10.1016/j.bbmt.2016.12.620] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/30/2016] [Accepted: 12/03/2016] [Indexed: 01/10/2023]
Abstract
The epidemiology of herpes zoster (HZ) in contemporary autologous hematopoietic cell transplant (HCT) recipients, and the impact of acyclovir (ACV)/valacyclovir (VACV) prophylaxis, is not well described. In this observational study from 2002 to 2010, we retrospectively identified 1000 varicella zoster virus (VZV)-seropositive autologous HCT recipients with up to 5 years of follow-up. The incidence of HZ and use of ACV/VACV prophylaxis were determined through review of medical records and mailed questionnaires. Risk factors for HZ were determined by multivariable Cox regression. Over a period of 5 years after autologous HCT, 194 patients developed at least 1 HZ episode, with a cumulative incidence of 21%; 159 of 194 (82%) were not on prophylaxis at the time of HZ. A second episode of HZ occurred in 31 of 194 (16%) patients. Patients taking ACV/VACV had reduced risk for HZ (adjusted hazard ratio [aHR], .59; 95% confidence interval [CI], .37 to .91), whereas those older than the median age (≥55.5 years) had increased risk (aHR, 1.42; 95% CI, 1.05 to 1.9). Disseminated VZV was reported in 8% and postherpetic neuralgia in 13% of patients. We demonstrate a high burden of HZ late after autologous HCT, despite long-term antiviral prophylaxis. Improved prevention strategies are needed to provide sustained protection against HZ after autologous HCT.
Collapse
Affiliation(s)
- Farah Sahoo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Public Health, University of Washington, Seattle, Washington
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Hu Xie
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Wendy Leisenring
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jessica Yi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sonia Goyal
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Louise E Kimball
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ingi Lee
- Merck and Co. Inc, Kenilworth, New Jersey
| | - Sachiko Seo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Chris Davis
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Stephen A Pergam
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington; Seattle Cancer Care Alliance, Seattle, Washington
| | - Mary E Flowers
- Department of Medicine, University of Washington, Seattle, Washington; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Leona Holmberg
- Department of Medicine, University of Washington, Seattle, Washington; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Seattle Cancer Care Alliance, Seattle, Washington
| | - Michael Boeckh
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington.
| |
Collapse
|
73
|
Brown AEC, Asturias EJ, Melgar M, Antillon-Klussmann FA, Mettler P, Levin MJ. Incidence and consequences of varicella in children treated for cancer in Guatemala. World J Pediatr 2016; 12:320-326. [PMID: 27351567 DOI: 10.1007/s12519-016-0025-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 02/24/2015] [Indexed: 01/04/2023]
Abstract
BACKGROUND Varicella-zoster virus infection is associated with significant morbidity and mortality in immune-compromised children, despite treatment with antiviral agents. Universal varicella vaccine programs have significantly decreased this risk in many highincome countries, but in most low-income and middleincome countries, the burden of varicella in children treated for malignancy is poorly defined. METHODS We retrospectively reviewed records of children at the National Unit of Pediatric Oncology (UNOP) in Guatemala diagnosed with varicella between January 2009 and March 2013 in order to calculate incidence of varicella and evaluate morbidity, mortality, treatment interruption, and cost. RESULTS Fifty-nine cases of varicella were identified. Incidence was 23.4 cases per 1000 person-years (p-y). 66.1% of cases occurred in children with leukemia (median age 5.2 years; interquantile range 3.4-7 years) and 41.0% of these occurred during maintenance therapy. Source of exposure was identified for 14/59 (23.7%) children. Most were hospitalized (71.2%) and given intravenous acyclovir (64.4%). Eight (13.6%) children required critical care, and two (3.4%) died from disseminated varicella with multiorgan failure. Chemotherapy was delayed or omitted due to varicella in 50%. No significant differences in outcomes based on nutritional and immunologic status were detected. The minimum average cost of treatment per episode was 598.75 USD. CONCLUSIONS Varicella is a significant problem in children treated for cancer in Guatemala, where effective post-exposure prophylaxis is limited. In the absence of universal varicella vaccination, strategies to improve recognition of exposure and the future use of novel inactivated vaccines currently under investigation in clinical trials could mitigate this burden.
Collapse
Affiliation(s)
- Amy E Caruso Brown
- Center for Bioethics and Humanities and Department of Pediatrics, SUNY Upstate Medical University, Syracuse, USA.
| | - Edwin J Asturias
- Center for Global Health and Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
| | - Mario Melgar
- National Unit of Pediatric Oncology, Guatemala City, Guatemala
| | | | - Pamela Mettler
- Department of Biostatistics and Informatics, Colorado Biostatistics Consortium, Colorado School of Public Health, Aurora, Colorado, USA
| | - Myron J Levin
- Section of Pediatric Infectious Diseases, Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
74
|
Fochesato M, Dendouga N, Boxus M. Comparative preclinical evaluation of AS01 versus other Adjuvant Systems in a candidate herpes zoster glycoprotein E subunit vaccine. Hum Vaccin Immunother 2016; 12:2092-2095. [PMID: 26933767 PMCID: PMC4994747 DOI: 10.1080/21645515.2016.1154247] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The candidate vaccine HZ/su is being developed to prevent herpes-zoster disease (HZ). HZ occurrence is attributed to declines in varicella-zoster virus (VZV) specific T-cell immunity. HZ/su contains VZV antigen, gE, and Adjuvant System AS01B (liposome-based formulation of MPL and QS-21). In clinical trials, AS01B enhances CD4+ T-cell responses to gE. In clinical trials of other vaccines, Adjuvant Systems AS03 and AS04 also enhance antigen-specific CD4+ T-cell responses. Hence the purpose of this study was to evaluate gE formulated with AS01B, AS01E (50% less MPL and QS-21 than AS01B), AS03 or AS04 in C57BL6 mice primed with live-attenuated VZV. Four-weeks post-vaccination, the gE-specific CD4+ T-cell response to gE/AS01B was 5.4, 2.8 and 2.2-fold greater than those to gE/AS03, gE/AS04 and gE/AS03, respectively (p<0.001). Therefore in the VZV-primed mouse model, CD4+ T-cell responses to gE appeared most enhanced by AS01B, and adds further support for the use of AS01B in the HZ/su formulation.
Collapse
Affiliation(s)
- Michel Fochesato
- a GSK Vaccines; Rue de l'Institut ; 89; 1330 ; Rixensart ; Belgium
| | - Najoua Dendouga
- a GSK Vaccines; Rue de l'Institut ; 89; 1330 ; Rixensart ; Belgium
| | - Mathieu Boxus
- a GSK Vaccines; Rue de l'Institut ; 89; 1330 ; Rixensart ; Belgium
| |
Collapse
|
75
|
Serological Evaluation of Immunity to the Varicella-Zoster Virus Based on a Novel Competitive Enzyme-Linked Immunosorbent Assay. Sci Rep 2016; 6:20577. [PMID: 26853741 PMCID: PMC4744930 DOI: 10.1038/srep20577] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/06/2016] [Indexed: 12/18/2022] Open
Abstract
Varicella-zoster virus (VZV) is a highly contagious agent of varicella and herpes zoster. Varicella can be lethal to immunocompromised patients, babies, HIV patients and other adults with impaired immunity. Serological evaluation of immunity to VZV will help determine which individuals are susceptible and evaluate vaccine effectiveness. A collection of 110 monoclonal antibodies (mAbs) were obtained by immunization of mice with membrane proteins or cell-free virus. The mAbs were well characterized, and a competitive sandwich ELISA (capture mAb: 8H6; labelling mAb: 1B11) was established to determine neutralizing antibodies in human serum with reference to the FAMA test. A total of 920 human sera were evaluated. The competitive sandwich ELISA showed a sensitivity of 95.6%, specificity of 99.77% and coincidence of 97.61% compared with the fluorescent-antibody-to-membrane-antigen (FAMA) test. The capture mAb 8H6 was characterized as a specific mAb for VZV ORF9, a membrane-associated tegument protein that interacts with glycoprotein E (gE), glycoprotein B (gB) and glycoprotein C (gC). The labelling mAb 1B11 was characterized as a complement-dependent neutralizing mAb specific for the immune-dominant epitope located on gE, not on other VZV glycoproteins. The established competitive sandwich ELISA could be used as a rapid and high-throughput method for evaluating immunity to VZV.
Collapse
|
76
|
Aerssens A, Leroux-Roels G. Adjuvanted herpes zoster subunit vaccine. Future Virol 2016. [DOI: 10.2217/fvl.15.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This review highlights the characteristics of a candidate herpes zoster (HZ) vaccine (HZ/su, GlaxoSmithKline Vaccines) that consists of 50 μg recombinant glycoprotein E (gE) of varicella zoster virus adjuvanted with AS01B. It is well tolerated and shows a clinically acceptable tolerability profile. It strongly enhances pre-existing gE-specific CD4+ T-cell and anti-gE antibody responses in older adults and immunocompromised persons. Administration of two doses 2 months apart reduces the risk of HZ by 97.2% in adults ≥50 years. This effect does not diminish with increasing age. Long-term persistence of its efficacy still needs to be determined. This candidate HZ vaccine may become an alternative for the high-dose live-attenuated varicella zoster virus vaccine for the prevention of HZ in older persons and in immunocompromised patients in whom the use of live-attenuated vaccines is contraindicated.
Collapse
Affiliation(s)
- Annelies Aerssens
- Centre for Vaccinology, Ghent University & Ghent University Hospital, Ghent, Belgium
| | - Geert Leroux-Roels
- Centre for Vaccinology, Ghent University & Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
77
|
Panatto D, Bragazzi NL, Rizzitelli E, Bonanni P, Boccalini S, Icardi G, Gasparini R, Amicizia D. Evaluation of the economic burden of Herpes Zoster (HZ) infection. Hum Vaccin Immunother 2015; 11:245-62. [PMID: 25483704 DOI: 10.4161/hv.36160] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The main objective of this systematic review was to evaluate the economic burden of Herpes Zoster (HZ) infection. The review was conducted in accordance with the standards of the "Preferred Reporting Items for Systematic Reviews and Meta-Analyses" guidelines. The following databases were accessed: ISI/Web of Knowledge (WoS), MEDLINE/PubMed, Scopus, ProQuest, the Cochrane Library and EconLit. Specific literature on health economics was also manually inspected. Thirty-three studies were included. The quality of the studies assessed in accordance with the Consolidated Health Economic Evaluation Reporting Standards checklist was good. All studies evaluated direct costs, apart from one which dealt only with indirect costs. Indirect costs were evaluated by 12 studies. The economic burden of HZ has increased over time. HZ management and drug prescriptions generate the highest direct costs. While increasing age, co-morbidities and drug treatment were found to predict higher direct costs, being employed was correlated with higher indirect costs, and thus with the onset age of the disease. Despite some differences among the selected studies, particularly with regard to indirect costs, all concur that HZ is a widespread disease which has a heavy social and economic burden.
Collapse
|
78
|
Purswani MU, Karalius B, Yao TJ, Schmid DS, Burchett SK, Siberry GK, Patel K, Van Dyke RB, Yogev R, Lurie RH, Yogev R, Sanders MA, Malee K, Hunter S, Shearer W, Paul M, Cooper N, Harris L, Purswani M, Baig M, Cintron A, Puga A, Navarro S, Garvie P, Blood J, Burchett S, Karthas N, Kammerer B, Wiznia A, Burey M, Nozyce M, Dieudonne A, Bettica L, Adubato S, Chen J, Bulkley MG, Ivey L, Grant M, Knapp K, Allison K, Wilkins M, Acevedo-Flores M, Rios H, Olivera V, Silio M, Jones M, Sirois P, Spector S, Norris K, Nichols S, McFarland E, Katai A, Dunn J, Paul S, Scott G, Bryan P, Willen E. Prevalence and Persistence of Varicella Antibodies in Previously Immunized Children and Youth With Perinatal HIV-1 Infection. Clin Infect Dis 2015; 62:106-114. [PMID: 26385992 DOI: 10.1093/cid/civ734] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 08/11/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Two doses of live-attenuated varicella-zoster vaccine are recommended for human immunodeficiency virus 1 (HIV-1)-infected children with CD4% ≥ 15%. We determined the prevalence and persistence of antibody in immunized children with perinatal HIV (PHIV) and their association with number of vaccinations, combination antiretroviral therapy (cART), and HIV status. METHODS The Adolescent Master Protocol is an observational study of children with PHIV and perinatally HIV-exposed but uninfected (PHEU) children conducted at 15 US sites. In a cross-sectional analysis, we tested participants' most recent stored sera for varicella antibody using whole-cell and glycoprotein enzyme-linked immunosorbent assay. Seropositivity predictors were identified using multivariable logistic regression models and C statistics. RESULTS Samples were available for 432 children with PHIV and 221 PHEU children; 82% of children with PHIV and 97% of PHEU children were seropositive (P < .001). Seropositivity after 1 vaccine dose among children with PHIV and PHEU children was 100% at <3 years (both), 73% and 100% at 3-<7 years (P < .05), and 77% and 97% at ≥ 7 years (P < .01), respectively. Seropositivity among recipients of 2 vaccine doses was >94% at all intervals. Independent predictors of seropositivity among children with PHIV were receipt of 2 vaccine doses, receipt of 1 dose while on ≥ 3 months of cART, compared with none (adjusted odds ratio [aOR]: 14.0 and 2.8, respectively; P < .001 for overall dose effect), and in those vaccinated ≥ 3 years previously, duration of cART (aOR: 1.29 per year increase, P = .02). CONCLUSIONS Humoral immune responses to varicella vaccine are best achieved when children with PHIV receive their first dose ≥ 3 months after cART initiation and maintained by completion of the 2-dose series and long-term cART use.
Collapse
Affiliation(s)
- Murli U Purswani
- Division of Pediatric Infectious Disease, Bronx-Lebanon Hospital Center, Albert Einstein College of Medicine, New York
| | - Brad Karalius
- Department of Epidemiology.,Center for Biostatistics in AIDS Research (CBAR), Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Tzy-Jyun Yao
- Center for Biostatistics in AIDS Research (CBAR), Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - D Scott Schmid
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Sandra K Burchett
- Boston Children's Hospital and Harvard Medical School, Massachusetts
| | - George K Siberry
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Kunjal Patel
- Department of Epidemiology.,Center for Biostatistics in AIDS Research (CBAR), Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Russell B Van Dyke
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ram Yogev
- Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Illinois
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Harrer A, Wipfler P, Pilz G, Oppermann K, Haschke-Becher E, Afazel S, Kraus J, Trinka E, Sellner J. Adaptive Immune Responses in a Multiple Sclerosis Patient with Acute Varicella-Zoster Virus Reactivation during Treatment with Fingolimod. Int J Mol Sci 2015; 16:21832-45. [PMID: 26378517 PMCID: PMC4613283 DOI: 10.3390/ijms160921832] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 11/22/2022] Open
Abstract
Fingolimod, an oral sphingosine 1-phosphate (S1P) receptor modulator, is approved for the treatment of relapsing forms of multiple sclerosis (MS). The interference with S1P signaling leads to retention particularly of chemokine receptor-7 (CCR7) expressing T cells in lymph nodes. The immunological basis of varicella zoster virus (VZV) infections during fingolimod treatment is unclear. Here, we studied the dynamics of systemic and intrathecal immune responses associated with symptomatic VZV reactivation including cessation of fingolimod and initiation of antiviral therapy. Key features in peripheral blood were an about two-fold increase of VZV-specific IgG at diagnosis of VZV reactivation as compared to the previous months, a relative enrichment of effector CD4+ T cells (36% versus mean 12% in controls), and an accelerated reconstitution of absolute lymphocytes counts including a normalized CD4+/CD8+ ratio and reappearance of CCR7+ T cells. In cerebrospinal fluid (CSF) the lymphocytic pleocytosis and CD4+/CD8+ ratios at diagnosis of reactivation and after nine days of fingolimod discontinuation remained unchanged. During this time CCR7+ T cells were not observed in CSF. Further research into fingolimod-associated VZV reactivation and immune reconstitution is mandatory to prevent morbidity and mortality associated with this potentially life-threatening condition.
Collapse
Affiliation(s)
- Andrea Harrer
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; E-Mails: (A.H.); (P.W.); (G.P.); (K.O.); (E.T.)
| | - Peter Wipfler
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; E-Mails: (A.H.); (P.W.); (G.P.); (K.O.); (E.T.)
| | - Georg Pilz
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; E-Mails: (A.H.); (P.W.); (G.P.); (K.O.); (E.T.)
| | - Katrin Oppermann
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; E-Mails: (A.H.); (P.W.); (G.P.); (K.O.); (E.T.)
| | - Elisabeth Haschke-Becher
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria; E-Mails: (E.H.-B.); (S.A.)
| | - Shahrzad Afazel
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria; E-Mails: (E.H.-B.); (S.A.)
| | - Jörg Kraus
- Department of Neurology, A.ö. Krankenhaus Zell am See, Teaching Hospital of the Paracelsus Medical University, 5700 Zell am See, Austria; E-Mail:
- Research Institute of Neurointervention, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Eugen Trinka
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; E-Mails: (A.H.); (P.W.); (G.P.); (K.O.); (E.T.)
| | - Johann Sellner
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020 Salzburg, Austria; E-Mails: (A.H.); (P.W.); (G.P.); (K.O.); (E.T.)
- Department of Neurology, Klinikum rechts der Isar, Technische Universität, 81675 München, Germany
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +43-662-4483-(ext. 0); Fax: +43-662-4483-3004
| |
Collapse
|
80
|
Vukmanovic-Stejic M, Sandhu D, Seidel JA, Patel N, Sobande TO, Agius E, Jackson SE, Fuentes-Duculan J, Suárez-Fariñas M, Mabbott NA, Lacy KE, Ogg G, Nestle FO, Krueger JG, Rustin MHA, Akbar AN. The Characterization of Varicella Zoster Virus-Specific T Cells in Skin and Blood during Aging. J Invest Dermatol 2015; 135:1752-1762. [PMID: 25734814 PMCID: PMC4471118 DOI: 10.1038/jid.2015.63] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 01/14/2015] [Accepted: 02/03/2015] [Indexed: 12/12/2022]
Abstract
Reactivation of the varicella zoster virus (VZV) increases during aging. Although the effects of VZV reactivation are observed in the skin (shingles), the number and functional capacity of cutaneous VZV-specific T cells have not been investigated. The numbers of circulating IFN-γ-secreting VZV-specific CD4(+) T cells are significantly decreased in old subjects. However, other measures of VZV-specific CD4(+) T cells, including proliferative capacity to VZV antigen stimulation and identification of VZV-specific CD4(+) T cells with an major histocompatibility complex class II tetramer (epitope of IE-63 protein), were similar in both age groups. The majority of T cells in the skin of both age groups expressed CD69, a characteristic of skin-resident T cells. VZV-specific CD4(+) T cells were significantly increased in the skin compared with the blood in young and old subjects, and their function was similar in both age groups. In contrast, the number of Foxp3(+) regulatory T cells and expression of the inhibitory receptor programmed cell death -1 PD-1 on CD4(+) T cells were significantly increased in the skin of older humans. Therefore, VZV-specific CD4(+) T cells in the skin of older individuals are functionally competent. However, their activity may be restricted by multiple inhibitory influences in situ.
Collapse
Affiliation(s)
| | - Daisy Sandhu
- Division of Infection and Immunity, University College London, London, UK; Department of Dermatology, Royal Free Hospital, London, UK
| | - Judith A Seidel
- Division of Infection and Immunity, University College London, London, UK
| | - Neil Patel
- Division of Infection and Immunity, University College London, London, UK; Department of Dermatology, Royal Free Hospital, London, UK
| | - Toni O Sobande
- Division of Infection and Immunity, University College London, London, UK
| | - Elaine Agius
- Division of Infection and Immunity, University College London, London, UK; Department of Dermatology, Royal Free Hospital, London, UK
| | - Sarah E Jackson
- Division of Infection and Immunity, University College London, London, UK
| | | | | | - Neil A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, UK
| | - Katie E Lacy
- St Johns Institute of Dermatology, Guys and St Thomas' Hospital, London, UK
| | - Graham Ogg
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Frank O Nestle
- St Johns Institute of Dermatology, Guys and St Thomas' Hospital, London, UK
| | - James G Krueger
- Laboratory for Investigative Dermatology, Rockefeller University, New York, USA
| | | | - Arne N Akbar
- Division of Infection and Immunity, University College London, London, UK.
| |
Collapse
|
81
|
Sarkadi J, Jankovics M, Fodor K, Kis Z, Takacs M, Visontai I, Jankovics I, Gonczol E. High-level cellular and humoral immune responses in Guinea pigs immunized intradermally with a heat-inactivated varicella-zoster virus vaccine. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:570-7. [PMID: 25787138 PMCID: PMC4412949 DOI: 10.1128/cvi.00773-14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/15/2015] [Indexed: 01/29/2023]
Abstract
The threat of varicella and herpes zoster in immunocompromised individuals necessitates the development of a safe and effective varicella-zoster virus (VZV) vaccine. The immune responses of guinea pigs to the intradermal (i.d.) or subcutaneous (s.c.) administration of a heat-inactivated or live VZV vaccine were investigated. Relative to nonimmunized animals, a single 399-PFU dose of vaccine induced nonsignificant increases in gamma interferon (IFN-γ), granzyme B, and perforin mRNA expression in the splenocytes of all groups, while two i.d. administrations of the inactivated vaccine increased IFN-γ mRNA expression significantly (P < 0.005). A single 1,995-PFU dose significantly increased the expression of IFN-γ mRNA in the groups receiving the vaccine either i.d. (P < 0.005) or s.c. (P < 0.05), that of granzyme B mRNA in the groups immunized i.d. with the inactivated (P < 0.005) or live (P < 0.005) vaccine, and that of perforin mRNA in the animals that received the inactivated vaccine i.d. (P < 0.005). Importantly, increases in the expression of IFN-γ (P = 0.025), granzyme B (P = 0.004), and perforin (P > 0.05) mRNAs were observed in the animals immunized i.d. with 1,995 PFU of inactivated vaccine relative to those immunized s.c. with the same dose. The proportion of animals expressing IFN-γ mRNA mirrored the proportion expressing IFN-γ protein (correlation coefficient of 0.88). VZV glycoprotein-specific and virus-neutralizing antibodies were produced with no significant intergroup differences. A booster i.d. administration of the 399-PFU dose of heat-inactivated vaccine enhanced the antibody responses. These results demonstrate that i.d. administration of an inactivated VZV vaccine can be an efficient mode of immunization against VZV.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Chickenpox Vaccine/administration & dosage
- Chickenpox Vaccine/immunology
- Granzymes/genetics
- Granzymes/metabolism
- Guinea Pigs
- Herpesvirus 3, Human/genetics
- Herpesvirus 3, Human/immunology
- Immunity, Cellular
- Immunity, Humoral
- Immunization, Secondary
- Injections, Intradermal
- Injections, Subcutaneous
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Perforin/genetics
- Perforin/immunology
- Spleen/cytology
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/immunology
- Vaccines, Inactivated/administration & dosage
- Vaccines, Inactivated/immunology
Collapse
Affiliation(s)
- Julia Sarkadi
- Division of Virology, National Center for Epidemiology, Budapest, Hungary
| | - Mate Jankovics
- Division of Virology, National Center for Epidemiology, Budapest, Hungary
| | - Kinga Fodor
- Faculty of Veterinary Science, Szent Istvan University, Budapest, Hungary
| | - Zoltan Kis
- Division of Virology, National Center for Epidemiology, Budapest, Hungary
| | - Maria Takacs
- Division of Virology, National Center for Epidemiology, Budapest, Hungary
| | - Ildiko Visontai
- Division of Virology, National Center for Epidemiology, Budapest, Hungary
| | - Istvan Jankovics
- Division of Virology, National Center for Epidemiology, Budapest, Hungary
| | - Eva Gonczol
- Division of Virology, National Center for Epidemiology, Budapest, Hungary
| |
Collapse
|
82
|
Laing KJ, Russell RM, Dong L, Schmid DS, Stern M, Magaret A, Haas JG, Johnston C, Wald A, Koelle DM. Zoster Vaccination Increases the Breadth of CD4+ T Cells Responsive to Varicella Zoster Virus. J Infect Dis 2015; 212:1022-31. [PMID: 25784732 DOI: 10.1093/infdis/jiv164] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 03/06/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The live, attenuated varicella vaccine strain (vOka) is the only licensed therapeutic vaccine. Boost of varicella zoster virus (VZV)-specific cellular immunity is a likely mechanism of action. We examined memory CD4(+) T-cell responses to each VZV protein at baseline and after zoster vaccination. METHODS Serial blood samples were collected from 12 subjects vaccinated with Zostavax and immunogenicity confirmed by ex vivo VZV-specific T-cell and antibody assays. CD4(+) T-cell lines enriched for VZV specificity were generated and probed for proliferative responses to every VZV protein and selected peptide sets. RESULTS Zoster vaccination increased the median magnitude (2.3-fold) and breadth (4.2-fold) of VZV-specific CD4(+) T cells one month post-vaccination. Both measures declined by 6 months. The most prevalent responses at baseline included VZV open reading frames (ORFs) 68, 4, 37, and 63. After vaccination, responses to ORFs 40, 67, 9, 59, 12, 62, and 18 were also prevalent. The immunogenicity of ORF9 and ORF18 were confirmed using peptides, defining a large number of discrete CD4 T-cell epitopes. CONCLUSIONS The breadth and magnitude of the VZV-specific CD4(+) T-cell response increase after zoster vaccination. In addition to glycoprotein E (ORF68), we identified antigenic ORFs that may be useful components of subunit vaccines.
Collapse
Affiliation(s)
- Kerry J Laing
- Department of Medicine, University of Washington, Seattle
| | | | - Lichun Dong
- Department of Medicine, University of Washington, Seattle
| | - D Scott Schmid
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | - Amalia Magaret
- Department of Laboratory Medicine Department of Biostatistics, University of Washington Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jürgen G Haas
- Division of Infection and Pathway Medicine, University of Edinburgh, United Kingdom
| | - Christine Johnston
- Department of Medicine, University of Washington, Seattle Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Anna Wald
- Department of Medicine, University of Washington, Seattle Department of Laboratory Medicine Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington Department of Epidemiology
| | - David M Koelle
- Department of Medicine, University of Washington, Seattle Department of Laboratory Medicine Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington Department of Global Health, University of Washington Benaroya Research Institute, Seattle, Washington
| |
Collapse
|
83
|
Hu Y, Smolkin ME, White EJ, Petroni GR, Neese PY, Slingluff CL. Inflammatory adverse events are associated with disease-free survival after vaccine therapy among patients with melanoma. Ann Surg Oncol 2014; 21:3978-84. [PMID: 24841355 PMCID: PMC4192070 DOI: 10.1245/s10434-014-3794-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Multipeptide vaccines for melanoma may cause inflammatory adverse events (IAE). We hypothesize that IAE are associated with a higher rate of immune response (IR) to vaccination and improved clinical outcomes. METHODS Adult patients with resected, high-risk (stage IIB to IV) melanoma were vaccinated with a combination of 12 class I major histocompatibility complex (MHC)-restricted melanoma epitopes, and IAE were recorded. A separate category for hypopigmentation (vitiligo) was also assessed. CD8(+) T cell IR was assessed by direct interferon gamma ELISpot analysis. Overall survival and disease-free survival were analyzed by Cox proportional hazard modeling. RESULTS Out of 332 patients, 57 developed IAE, the majority of which were dermatologic (minimum Common Terminology Criteria for Adverse Events [CTCAE] grade 3). Most nondermatologic IAE were CTCAE grade 1 and 2. Vitiligo developed in 23 patients (7 %). A total of 174 patients (53 %) developed a CD8(+) response. Presence of IAE was significantly associated with development of IR (70 vs. 49 %, p = 0.005) and with disease-free survival (hazard ratio 0.54, p = 0.043). There were no significant associations relating vitiligo or IR alone with clinical outcomes. CONCLUSIONS IAE are associated with a higher rate of CD8(+) T cell response after vaccination therapy for high-risk melanoma. Our findings suggest either that antitumor activity induced by class I MHC-restricted peptide vaccines may depend on immunologic effects beyond simple expansion of CD8(+) T cells or that the intrinsic inflammatory response of patients contributes to clinical outcome in melanoma.
Collapse
Affiliation(s)
- Yinin Hu
- Department of Surgery/Division of Surgical Oncology, University of Virginia Health System, Charlottesville, VA, USA,
| | | | | | | | | | | |
Collapse
|
84
|
Van Epps P, Banks R, Aung H, Betts MR, Canaday DH. Age-related differences in polyfunctional T cell responses. IMMUNITY & AGEING 2014; 11:14. [PMID: 25512758 PMCID: PMC4265991 DOI: 10.1186/1742-4933-11-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 10/09/2014] [Indexed: 12/21/2022]
Abstract
Background A reduced number of naïve T cells along with an accumulation of differentiated cell types in aging have been described but little is known about the polyfunctionality of the T cell responses. In this study we compared the individual and polyfunctional expression of IFN-γ, MIP-1α, TNF-α, perforin, and IL-2 by T cell subsets, including the newly described stem cell like memory T cells (TSCM), in response to stimulation with superantigen staphylococcal enterotoxin B (SEB) in older (median age 80, n = 23) versus younger (median age 27; n = 23) adults. Results Older age was associated with a markedly lower frequency of CD8+ naïve T cells (11% vs. 47%; p < 0.0001) and an expansion in memory T cell subsets including central memory (p < 0.05), effector memory and effector T cells (p < 0.001 for both). There was also a decline in CD4+ naïve T cells in older subjects (33% vs. 45%; p = 0.02). There were no differences in frequencies or polyfunctional profiles of TSCM between groups. CD8+ naïve cells in the older group had increased expression of all functional parameters measured compared to the younger subjects and exhibited greater polyfunctionality (p = 0.04). CD4+ naïve T cells in the older group also showed greater polyfunctionality with a TNF-α and IL-2 predominance (p = 0.005). CD8+ effector memory and effector T cells exhibited increased polyfunctionality in the older group compared with younger (p = 0.01 and p = 0.003). Conclusions These data suggest that aging does not have a negative effect on polyfunctionality and therefore this is likely not a major contributor to the immunesenescence described with aging.
Collapse
Affiliation(s)
- Puja Van Epps
- Geriatric Research Center Clinical Core (GRECC), Louis Stokes Cleveland VA Medical Center, 10701 East Blvd, Cleveland, Ohio 44106, USA ; Division of Infectious Diseases, Case Western Reserve University School of Medicine, 10900 Euclid Ave, BRB 1022, Cleveland, Ohio, 44106-4684, USA
| | - Richard Banks
- Geriatric Research Center Clinical Core (GRECC), Louis Stokes Cleveland VA Medical Center, 10701 East Blvd, Cleveland, Ohio 44106, USA
| | - Htin Aung
- Geriatric Research Center Clinical Core (GRECC), Louis Stokes Cleveland VA Medical Center, 10701 East Blvd, Cleveland, Ohio 44106, USA ; Division of Infectious Diseases, Case Western Reserve University School of Medicine, 10900 Euclid Ave, BRB 1022, Cleveland, Ohio, 44106-4684, USA
| | - Michael R Betts
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - David H Canaday
- Geriatric Research Center Clinical Core (GRECC), Louis Stokes Cleveland VA Medical Center, 10701 East Blvd, Cleveland, Ohio 44106, USA ; Division of Infectious Diseases, Case Western Reserve University School of Medicine, 10900 Euclid Ave, BRB 1022, Cleveland, Ohio, 44106-4684, USA
| |
Collapse
|
85
|
Sen N, Mukherjee G, Sen A, Bendall SC, Sung P, Nolan GP, Arvin AM. Single-cell mass cytometry analysis of human tonsil T cell remodeling by varicella zoster virus. Cell Rep 2014; 8:633-45. [PMID: 25043183 PMCID: PMC4127309 DOI: 10.1016/j.celrep.2014.06.024] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 05/17/2014] [Accepted: 06/17/2014] [Indexed: 12/11/2022] Open
Abstract
Although pathogens must infect differentiated host cells that exhibit substantial diversity, documenting the consequences of infection against this heterogeneity is challenging. Single-cell mass cytometry permits deep profiling based on combinatorial expression of surface and intracellular proteins. We used this method to investigate varicella-zoster virus (VZV) infection of tonsil T cells, which mediate viral transport to skin. Our results indicate that VZV induces a continuum of changes regardless of basal phenotypic and functional T cell characteristics. Contrary to the premise that VZV selectively infects T cells with skin trafficking profiles, VZV infection altered T cell surface proteins to enhance or induce these properties. Zap70 and Akt signaling pathways that trigger such surface changes were activated in VZV-infected naive and memory cells by a T cell receptor (TCR)-independent process. Single-cell mass cytometry is likely to be broadly relevant for demonstrating how intracellular pathogens modulate differentiated cells to support pathogenesis in the natural host.
Collapse
Affiliation(s)
- Nandini Sen
- Department of Pediatrics, Stanford University, Stanford, CA 94025, USA
| | - Gourab Mukherjee
- Department of Statistics, Stanford University, Stanford, CA 94025, USA; Department of Data Sciences and Operations, University of Southern California, Los Angeles, CA 90089, USA
| | - Adrish Sen
- Department of Medicine, Stanford University, Stanford, CA 94025, USA
| | - Sean C Bendall
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94025, USA
| | - Phillip Sung
- Department of Pediatrics, Stanford University, Stanford, CA 94025, USA
| | - Garry P Nolan
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94025, USA
| | - Ann M Arvin
- Department of Pediatrics, Stanford University, Stanford, CA 94025, USA; Department of Microbiology and Immunology, Stanford University, Stanford, CA 94025, USA.
| |
Collapse
|
86
|
Papaloukas O, Giannouli G, Papaevangelou V. Successes and challenges in varicella vaccine. THERAPEUTIC ADVANCES IN VACCINES 2014; 2:39-55. [PMID: 24757524 DOI: 10.1177/2051013613515621] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Varicella is a highly contagious disease caused by primary infection with varicella zoster virus (VZV). VZV infection, as well as varicella vaccination, induces VZV-specific antibody and T-cell-mediated immunity, essential for recovery. The immune responses developed contribute to protection following re-exposure to VZV. When cell-mediated immunity declines, as occurs with aging or immunosuppression, reactivation of VZV leads to herpes zoster (HZ). It has been almost 20 years since universal varicella vaccination has been implemented in many areas around the globe and this has resulted in a significant reduction of varicella-associated disease burden. Successes are reviewed here, whilst emphasis is put on the challenges ahead. Most countries that have not implemented routine childhood varicella vaccination have chosen to vaccinate high-risk groups alone. The main reasons for not introducing universal vaccination are discussed, including fear of age shift of peak incidence age and of HZ incidence increase. Possible reasons for not observing the predicted increase in HZ incidence are explored. The advantages and disadvantages of universal vs targeted vaccination as well as different vaccination schedules are discussed.
Collapse
Affiliation(s)
- Orestis Papaloukas
- Second Department of Pediatrics, University of Athens Medical School, P&A Kyriakou Childrens' Hospital, Greece
| | - Georgia Giannouli
- Second Department of Pediatrics, University of Athens Medical School, P&A Kyriakou Childrens' Hospital, Greece
| | - Vassiliki Papaevangelou
- Third Department of Pediatrics, University of Athens Medical School, General University Hospital 'ATTIKON', Rimini 1, Chaidari 124 62, Greece
| |
Collapse
|
87
|
Chiu C, McCausland M, Sidney J, Duh FM, Rouphael N, Mehta A, Mulligan M, Carrington M, Wieland A, Sullivan NL, Weinberg A, Levin MJ, Pulendran B, Peters B, Sette A, Ahmed R. Broadly reactive human CD8 T cells that recognize an epitope conserved between VZV, HSV and EBV. PLoS Pathog 2014; 10:e1004008. [PMID: 24675761 PMCID: PMC3968128 DOI: 10.1371/journal.ppat.1004008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 02/03/2014] [Indexed: 02/06/2023] Open
Abstract
Human herpesviruses are important causes of potentially severe chronic infections for which T cells are believed to be necessary for control. In order to examine the role of virus-specific CD8 T cells against Varicella Zoster Virus (VZV), we generated a comprehensive panel of potential epitopes predicted in silico and screened for T cell responses in healthy VZV seropositive donors. We identified a dominant HLA-A*0201-restricted epitope in the VZV ribonucleotide reductase subunit 2 and used a tetramer to analyze the phenotype and function of epitope-specific CD8 T cells. Interestingly, CD8 T cells responding to this VZV epitope also recognized homologous epitopes, not only in the other α-herpesviruses, HSV-1 and HSV-2, but also the γ-herpesvirus, EBV. Responses against these epitopes did not depend on previous infection with the originating virus, thus indicating the cross-reactive nature of this T cell population. Between individuals, the cells demonstrated marked phenotypic heterogeneity. This was associated with differences in functional capacity related to increased inhibitory receptor expression (including PD-1) along with decreased expression of co-stimulatory molecules that potentially reflected their stimulation history. Vaccination with the live attenuated Zostavax vaccine did not efficiently stimulate a proliferative response in this epitope-specific population. Thus, we identified a human CD8 T cell epitope that is conserved in four clinically important herpesviruses but that was poorly boosted by the current adult VZV vaccine. We discuss the concept of a "pan-herpesvirus" vaccine that this discovery raises and the hurdles that may need to be overcome in order to achieve this.
Collapse
Affiliation(s)
- Christopher Chiu
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Centre for Respiratory Infection, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Megan McCausland
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Fuh-Mei Duh
- Cancer and Inflammation Program, Laboratory for Experimental Immunology, SAIC Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Division of Infectious Diseases School of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Aneesh Mehta
- Division of Infectious Diseases School of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Mark Mulligan
- Hope Clinic of the Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Division of Infectious Diseases School of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Mary Carrington
- Cancer and Inflammation Program, Laboratory for Experimental Immunology, SAIC Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Andreas Wieland
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Nicole L. Sullivan
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Adriana Weinberg
- Departments of Pediatrics, Medicine and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Myron J. Levin
- Departments of Pediatrics, Medicine and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Bali Pulendran
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
88
|
Walton S, Mandaric S, Oxenius A. CD4 T cell responses in latent and chronic viral infections. Front Immunol 2013; 4:105. [PMID: 23717308 PMCID: PMC3651995 DOI: 10.3389/fimmu.2013.00105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 04/22/2013] [Indexed: 12/24/2022] Open
Abstract
The spectrum of tasks which is fulfilled by CD4 T cells in the setting of viral infections is large, ranging from support of CD8 T cells and humoral immunity to exertion of direct antiviral effector functions. While our knowledge about the differentiation pathways, plasticity, and memory of CD4 T cell responses upon acute infections or immunizations has significantly increased during the past years, much less is still known about CD4 T cell differentiation and their beneficial or pathological functions during persistent viral infections. In this review we summarize current knowledge about the differentiation, direct or indirect antiviral effector functions, and the regulation of virus-specific CD4 T cells in the setting of persistent latent or active chronic viral infections with a particular emphasis on herpes virus infections for the former and chronic lymphocytic choriomeningitis virus infection for the latter.
Collapse
Affiliation(s)
- Senta Walton
- Department of Microbiology and Immunology, School of Pathology and Laboratory Medicine, University of Western Australia Nedlands, WA, Australia
| | | | | |
Collapse
|
89
|
Rajakulendran S, Pathak S, Tai YF, Sanderson F, Davies NWS. VZV uveoretino meningitis following dental treatment. J Neurovirol 2013; 19:188-9. [PMID: 23508283 DOI: 10.1007/s13365-013-0159-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 02/09/2013] [Accepted: 02/25/2013] [Indexed: 11/24/2022]
Affiliation(s)
- Sanjeev Rajakulendran
- Charing Cross Hospital, Imperial College NHS Healthcare Trust, Fulham Palace Road, London, W6 8RF, UK
| | | | | | | | | |
Collapse
|
90
|
Irwin MR, Levin MJ, Laudenslager ML, Olmstead R, Lucko A, Lang N, Carrillo C, Stanley HA, Caulfield MJ, Weinberg A, Chan ISF, Clair J, Smith JG, Marchese RD, Williams HM, Beck DJ, McCook PT, Zhang JH, Johnson G, Oxman MN. Varicella zoster virus-specific immune responses to a herpes zoster vaccine in elderly recipients with major depression and the impact of antidepressant medications. Clin Infect Dis 2013; 56:1085-93. [PMID: 23413415 DOI: 10.1093/cid/cis1208] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The Depression Substudy of the Shingles Prevention Study (SPS) was designed to evaluate the association between major depression and immune responses to a high-titer live attenuated varicella zoster virus (VZV) vaccine (zoster vaccine), which boosts cell-mediated immunity (CMI) to VZV and decreases the incidence and severity of herpes zoster (HZ). The Depression Substudy was a 2-year longitudinal cohort study in 92 community-dwelling adults≥60 years of age who were enrolled in the SPS, a large, double-blind, placebo-controlled Veterans Affairs Cooperative zoster vaccine efficacy study. METHODS Forty subjects with major depressive disorder, stratified by use of antidepressant medications, and 52 age- and sex-matched controls with no history of depression or other mental illness had their VZV-CMI measured prior to vaccination with zoster vaccine or placebo and at 6 weeks, 1 year, and 2 years postvaccination. RESULTS Depressed subjects who were not treated with antidepressant medications had lower levels of VZV-CMI following administration of zoster vaccine than nondepressed controls or depressed subjects receiving antidepressants even when antidepressant medications failed to alter depressive symptom severity (P<.005). Similar results were obtained taking into account the time-varying status of depression and use of antidepressant medications, as well as changes in depressive symptoms, during the postvaccination period. CONCLUSIONS Depressed patients have diminished VZV-CMI responses to zoster vaccine, and treatment with antidepressant medication is associated with normalization of these responses. Because higher levels of VZV-CMI correlate with lower risk and severity of HZ, untreated depression may increase the risk and severity of HZ and reduce the efficacy of zoster vaccine.
Collapse
Affiliation(s)
- Michael R Irwin
- Cousins Center for Psychoneuroimmunology, UCLA Semel Institute for Neuroscience, University of California, Los Angeles, Los Angeles, CA 90095-7057, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
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: 3.7] [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
|
92
|
Vukmanovic-Stejic M, Sandhu D, Sobande TO, Agius E, Lacy KE, Riddell N, Montez S, Dintwe OB, Scriba TJ, Breuer J, Nikolich-Zugich J, Ogg G, Rustin MH, Akbar AN. Varicella zoster-specific CD4+Foxp3+ T cells accumulate after cutaneous antigen challenge in humans. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:977-86. [PMID: 23284056 PMCID: PMC3552094 DOI: 10.4049/jimmunol.1201331] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We investigated the relationship between varicella zoster virus (VZV)-specific memory CD4(+) T cells and CD4(+)Foxp3(+) regulatory T cells (Tregs) that accumulate after intradermal challenge with a VZV skin test Ag. VZV-specific CD4(+) T cells were identified with a MHC class II tetramer or by intracellular staining for either IFN-γ or IL-2 after Ag rechallenge in vitro. VZV-specific T cells, mainly of a central memory (CD45RA(-)CD27(+)) phenotype, accumulate at the site of skin challenge compared with the blood of the same individuals. This resulted in part from local proliferation because >50% of tetramer defined Ag-specific CD4(+) T cells in the skin expressed the cell cycle marker Ki67. CD4(+)Foxp3(+) T cells had the characteristic phenotype of Tregs, namely CD25(hi)CD127(lo)CD39(hi) in both unchallenged and VZV challenged skin and did not secrete IFN-γ or IL-2 after antigenic restimulation. The CD4(+)Foxp3(+) T cells from unchallenged skin had suppressive activity, because their removal led to an increase in cytokine secretion after activation. After VZV Ag injection, Foxp3(+)CD25(hi)CD127(lo)CD39(hi) T cells were also found within the VZV tetramer population. Their suppressive activity could not be directly assessed by CD25 depletion because activated T cells in the skin were also CD25(+). Nevertheless, there was an inverse correlation between decreased VZV skin responses and proportion of CD4(+)Foxp3(+) T cells present, indicating indirectly their inhibitory activity in vivo. These results suggest a linkage between the expansion of Ag-specific CD4(+) T cells and CD4(+) Tregs that may provide controlled responsiveness during Ag-specific stimulation in tissues.
Collapse
Affiliation(s)
- Milica Vukmanovic-Stejic
- Division of Infection and Immunity, University College London, London, W1T 4JF, England, United Kingdom
| | - Daisy Sandhu
- Division of Infection and Immunity, University College London, London, W1T 4JF, England, United Kingdom
- Department of Dermatology, Royal Free Hospital, London, NW3 2QG, England, United Kingdom
| | - Toni O. Sobande
- Division of Infection and Immunity, University College London, London, W1T 4JF, England, United Kingdom
| | - Elaine Agius
- Division of Infection and Immunity, University College London, London, W1T 4JF, England, United Kingdom
- Department of Dermatology, Royal Free Hospital, London, NW3 2QG, England, United Kingdom
| | - Katie E. Lacy
- Division of Infection and Immunity, University College London, London, W1T 4JF, England, United Kingdom
- Department of Dermatology, Royal Free Hospital, London, NW3 2QG, England, United Kingdom
- NIHR Biomedical Research Centre at Guy’s and St. Thomas’s Hospitals and King’s College London, Cutaneous Medicine and Immunotherapy, St. John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, King’s College London, London, UK
| | - Natalie Riddell
- Division of Infection and Immunity, University College London, London, W1T 4JF, England, United Kingdom
| | - Sandra Montez
- Division of Infection and Immunity, University College London, London, W1T 4JF, England, United Kingdom
| | - One B. Dintwe
- South African Tuberculosis Vaccine Initiative and School of Child and Adolescent Health, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative and School of Child and Adolescent Health, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Judith Breuer
- Division of Infection and Immunity, University College London, London, W1T 4JF, England, United Kingdom
| | - Janko Nikolich-Zugich
- Department of Immunobiology and the Arizona Center on Aging, University of Arizona College of Medicine, Tucson, AZ, 85719, USA
| | - Graham Ogg
- MRC Human Immunology Unit, University of Oxford, NIHR Biomedical Research Centre, Oxford, UK
| | - Malcolm H.A. Rustin
- Department of Dermatology, Royal Free Hospital, London, NW3 2QG, England, United Kingdom
| | - Arne N. Akbar
- Division of Infection and Immunity, University College London, London, W1T 4JF, England, United Kingdom
| |
Collapse
|
93
|
|
94
|
|
95
|
Lee KS, Zhou W, Scott-McKean JJ, Emmerling KL, Cai GY, Krah DL, Costa AC, Freed CR, Levin MJ. Human sensory neurons derived from induced pluripotent stem cells support varicella-zoster virus infection. PLoS One 2012; 7:e53010. [PMID: 23285249 PMCID: PMC3532467 DOI: 10.1371/journal.pone.0053010] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 11/26/2012] [Indexed: 12/16/2022] Open
Abstract
After primary infection, varicella-zoster virus (VZV) establishes latency in neurons of the dorsal root and trigeminal ganglia. Many questions concerning the mechanism of VZV pathogenesis remain unanswered, due in part to the strict host tropism and inconsistent availability of human tissue obtained from autopsies and abortions. The recent development of induced pluripotent stem (iPS) cells provides great potential for the study of many diseases. We previously generated human iPS cells from skin fibroblasts by introducing four reprogramming genes with non-integrating adenovirus. In this study, we developed a novel protocol to generate sensory neurons from iPS cells. Human iPS cells were exposed to small molecule inhibitors for 10 days, which efficiently converted pluripotent cells into neural progenitor cells (NPCs). The NPCs were then exposed for two weeks to growth factors required for their conversion to sensory neurons. The iPS cell-derived sensory neurons were characterized by immunocytochemistry, flow cytometry, RT-qPCR, and electrophysiology. After differentiation, approximately 80% of the total cell population expressed the neuron-specific protein, βIII-tubulin. Importantly, 15% of the total cell population co-expressed the markers Brn3a and peripherin, indicating that these cells are sensory neurons. These sensory neurons could be infected by both VZV and herpes simplex virus (HSV), a related alphaherpesvirus. Since limited neuronal populations are capable of supporting the entire VZV and HSV life cycles, our iPS-derived sensory neuron model may prove useful for studying alphaherpesvirus latency and reactivation.
Collapse
Affiliation(s)
- Katherine S Lee
- Department of Pediatrics, Section of Infectious Diseases, University of Colorado Denver, Aurora, Colorado, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Abstract
Varicella zoster virus (VZV) is a highly successful human pathogen, which is never completely eliminated from the host. VZV causes two clinically distinct diseases, varicella (chickenpox) during primary infection and herpes zoster (shingles) following virus reactivation from latency. Throughout its lifecycle the virus encounters the innate and adaptive immune response, and in order to prevent eradication it has developed many mechanisms to evade and overcome these responses. This review will provide a comprehensive overview of the host immune response to VZV infection, during the multiple stages of the virus lifecycle and at key sites of VZV infection. We will also briefly describe some of the strategies employed by the virus to overcome the host immune response and the ongoing challenges in further elucidating the interplay between VZV and the host immune response in an attempt to lead to better therapies and a ‘second generation’ vaccine for VZV disease.
Collapse
Affiliation(s)
- Megan Steain
- Discipline of Infectious Diseases & Immunology, The University of Sydney, NSW, Australia
- Centre for Virus Research, Westmead Millennium Institute, NSW, Australia
| | - Barry Slobedman
- Discipline of Infectious Diseases & Immunology, The University of Sydney, NSW, Australia
- Centre for Virus Research, Westmead Millennium Institute, NSW, Australia
| | - Allison Abendroth
- Discipline of Infectious Diseases & Immunology, The University of Sydney, NSW, Australia
| |
Collapse
|
97
|
Madan RP, Herold BC. Mounting evidence suggests safety and efficacy of immunizations posttransplantation. Am J Transplant 2012; 12:2871-2. [PMID: 23107269 DOI: 10.1111/j.1600-6143.2012.04276.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
98
|
COMMENTARY: Significantly less anti-gC antibody detectable in sera collected after varicella vaccination than after the disease varicella. Pediatr Infect Dis J 2012; 31:1153-4. [PMID: 23069796 PMCID: PMC3474975 DOI: 10.1097/inf.0b013e31826ef456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Varicella-zoster virus (VZV) is the first human herpesvirus to be attenuated and then approved in 1995 as a live vaccine for children. Within a few years after its administration in the United States, small outbreaks of breakthrough varicella were observed in vaccinees. Several risk factors were determined. But now a new investigation suggests another risk factor, namely, a deficiency in antibody responses to a specific individual VZV glycoprotein called gC (ORF14; gpV) in the vaccinees. Antibody concentrations to 5 VZV protein antigens were measured in children who had either wild type varicella or varicella vaccination. These proteins included two major glycoproteins called gE (ORF68; gpI) and gC (ORF14), both constituents of the viral envelope and therefore potentially important targets of the adaptive immune response. Of particular interest, the serum antibody responses to VZV gC antigen were significantly lower in vaccinees than in children who had wild type varicella. In contrast, the serum antibody responses to VZV gE antigen were comparable in both groups. These data implied that relatively little gC antigen was produced in children who were immunized. Since abundant gC protein is produced in skin vesicles during wild type varicella, the lack of a vesicular rash after vaccination may limit the amounts of some viral antigens required for an optimal antibody response.
Collapse
|
99
|
Levin MJ. Immune senescence and vaccines to prevent herpes zoster in older persons. Curr Opin Immunol 2012; 24:494-500. [PMID: 22857823 DOI: 10.1016/j.coi.2012.06.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 06/01/2012] [Accepted: 06/07/2012] [Indexed: 10/28/2022]
Abstract
Varicella-zoster virus (VZV) T-cell-mediated immunity (VZV-CMI) in older persons prevents latent VZV in sensory neurons from reactivating to cause herpes zoster. VZV-CMI declines greatly with aging, but can be restored by the licensed zoster vaccine. However, the vaccine-induced boost in VZV-CMI (which determines the efficacy of the vaccine) is a function of the age of the vaccinee, and the duration of this boost wanes with time. Both factors influence the value of this vaccine. To understand these aging effects, limited information about the phenotypic and functional differences in VZV-CMI in old and young persons are reviewed, as well as the reversal of these differences by vaccination. Based on information from these studies some potential approaches to improving prevention of herpes zoster are discussed.
Collapse
Affiliation(s)
- Myron J Levin
- Section of Pediatric Infectious Diseases, University of Colorado Denver School of Medicine and Health Sciences Center, C227, Building 401, 1784 Racine Street, Aurora, CO 80045, USA.
| |
Collapse
|
100
|
Affiliation(s)
- Marion Roderick
- Department of Paediatric Rheumatology, Bristol Royal Hospital for Children, 23 Hillview, Henleaze, Bristol BS9 4QD, UK
| | | | | |
Collapse
|