1
|
Scalabrin S, Becco A, Vitale A, Nuzzi R. Ocular effects caused by viral infections and corresponding vaccines: An overview of varicella zoster virus, measles virus, influenza viruses, hepatitis B virus, and SARS-CoV-2. Front Med (Lausanne) 2022; 9:999251. [PMID: 36388944 PMCID: PMC9643669 DOI: 10.3389/fmed.2022.999251] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/30/2022] [Indexed: 09/19/2023] Open
Abstract
Many viral infections can affect vision and the visual system. Vaccination to prevent diseases is commonplace today, acting by stimulating an immune response without developing the pathology. It involves the production of persisting antibodies against the pathogen and the activation of T cells. Certain diseases have already been eradicated by rigorous vaccination campaigns, while others are hoped to be eliminated soon. Vaccines currently available on the market are largely safe, even if they can rarely cause some adverse effects, such as ocular complications. Analyzing existing literature, we aimed to compare the pathological effects on the eye due to the most common viral infections [in particular varicella zoster virus (VZV), measles virus, influenza viruses, hepatitis B virus, and SARS-CoV-2] with the possible ocular adverse effects of their relative vaccines, in order to establish a risk-benefit relationship from an ophthalmological point of view.
Collapse
Affiliation(s)
| | | | | | - Raffaele Nuzzi
- Department of Surgical Sciences, Eye Clinic, University of Turin, Turin, Italy
| |
Collapse
|
2
|
Wu BW, Yee MB, Goldstein RS, Kinchington PR. Antiviral Targeting of Varicella Zoster Virus Replication and Neuronal Reactivation Using CRISPR/Cas9 Cleavage of the Duplicated Open Reading Frames 62/71. Viruses 2022; 14:v14020378. [PMID: 35215971 PMCID: PMC8880005 DOI: 10.3390/v14020378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 12/29/2022] Open
Abstract
Varicella Zoster Virus (VZV) causes Herpes Zoster (HZ), a common debilitating and complicated disease affecting up to a third of unvaccinated populations. Novel antiviral treatments for VZV reactivation and HZ are still in need. Here, we evaluated the potential of targeting the replicating and reactivating VZV genome using Clustered Regularly Interspaced Short Palindromic Repeat-Cas9 nucleases (CRISPR/Cas9) delivered by adeno-associated virus (AAV) vectors. After AAV serotype and guide RNA (gRNA) optimization, we report that a single treatment with AAV2-expressing Staphylococcus aureus CRISPR/Cas9 (saCas9) with gRNA to the duplicated and essential VZV genes ORF62/71 (AAV2-62gRsaCas9) greatly reduced VZV progeny yield and cell-to-cell spread in representative epithelial cells and in lytically infected human embryonic stem cell (hESC)-derived neurons. In contrast, AAV2-62gRsaCas9 did not reduce the replication of a recombinant virus mutated in the ORF62 targeted sequence, establishing that antiviral effects were a consequence of VZV-genome targeting. Delivery to latently infected and reactivation-induced neuron cultures also greatly reduced infectious-virus production. These results demonstrate the potential of AAV-delivered genome editors to limit VZV productive replication in epithelial cells, infected human neurons, and upon reactivation. The approach could be developed into a strategy for the treatment of VZV disease and virus spread in HZ.
Collapse
Affiliation(s)
- Betty W. Wu
- Graduate Program in Microbiology and Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
- Department of Ophthalmology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Michael B. Yee
- Department of Ophthalmology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | | | - Paul R. Kinchington
- Department of Ophthalmology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
- Correspondence: ; Tel.: +1-412-647-6319
| |
Collapse
|
3
|
Vaccination for quality of life: herpes-zoster vaccines. Aging Clin Exp Res 2021; 33:1113-1122. [PMID: 31643072 DOI: 10.1007/s40520-019-01374-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 10/04/2019] [Indexed: 12/14/2022]
Abstract
Current vaccination policy in most high-income countries aims to counteract the decline in cell-mediated immunity to varicella zoster virus that occurs with advancing age or immunosuppression. The aim of this review was to describe the burden of illness associated with herpes zoster (HZ) and post-herpetic neuralgia (PHN) risks and their impact on the social and common life in infected people. The effectiveness/efficacy and cost effectiveness of the immunization strategy will be presented through the review of the literature relevant to the live attenuated HZ vaccine (ZLV) licensed in 2006 and the recombinant HZ vaccine (RZV). The latter has very recently been approved to protect aged people aged ≥ 50 years against HZ morbidity including its complications, and associated health-care costs. Finally, this review also provides data with respect of precautions of using and safety of ZVL and RVZ.
Collapse
|
4
|
Advances and Perspectives in the Management of Varicella-Zoster Virus Infections. Molecules 2021; 26:molecules26041132. [PMID: 33672709 PMCID: PMC7924330 DOI: 10.3390/molecules26041132] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Varicella-zoster virus (VZV), a common and ubiquitous human-restricted pathogen, causes a primary infection (varicella or chickenpox) followed by establishment of latency in sensory ganglia. The virus can reactivate, causing herpes zoster (HZ, shingles) and leading to significant morbidity but rarely mortality, although in immunocompromised hosts, VZV can cause severe disseminated and occasionally fatal disease. We discuss VZV diseases and the decrease in their incidence due to the introduction of live-attenuated vaccines to prevent varicella or HZ. We also focus on acyclovir, valacyclovir, and famciclovir (FDA approved drugs to treat VZV infections), brivudine (used in some European countries) and amenamevir (a helicase-primase inhibitor, approved in Japan) that augur the beginning of a new era of anti-VZV therapy. Valnivudine hydrochloride (FV-100) and valomaciclovir stearate (in advanced stage of development) and several new molecules potentially good as anti-VZV candidates described during the last year are examined. We reflect on the role of antiviral agents in the treatment of VZV-associated diseases, as a large percentage of the at-risk population is not immunized, and on the limitations of currently FDA-approved anti-VZV drugs. Their low efficacy in controlling HZ pain and post-herpetic neuralgia development, and the need of multiple dosing regimens requiring daily dose adaptation for patients with renal failure urges the development of novel anti-VZV drugs.
Collapse
|
5
|
Hughes CM, Liu L, Davidson WB, Radford KW, Wilkins K, Monroe B, Metcalfe MG, Likafi T, Lushima RS, Kabamba J, Nguete B, Malekani J, Pukuta E, Karhemere S, Muyembe Tamfum JJ, Okitolonda Wemakoy E, Reynolds MG, Schmid DS, McCollum AM. A Tale of Two Viruses: Coinfections of Monkeypox and Varicella Zoster Virus in the Democratic Republic of Congo. Am J Trop Med Hyg 2020. [PMID: 33289470 DOI: 10.4269/ajtmh.200589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Recent enhanced monkeypox (MPX) surveillance in the Democratic Republic of Congo, where MPX is endemic, has uncovered multiple cases of MPX and varicella zoster virus (VZV) coinfections. The purpose of this study was to verify if coinfections occur and to characterize the clinical nature of these cases. Clinical, epidemiological, and laboratory results were used to investigate MPX/VZV coinfections. A coinfection was defined as a patient with at least one Orthopoxvirus/MPX-positive sample and at least one VZV-positive sample within the same disease event. Between September 2009 and April 2014, 134 of the 1,107 (12.1%) suspected MPX cases were confirmed as MPX/VZV coinfections. Coinfections were more likely to report symptoms than VZV-alone cases and less likely than MPX-alone cases. Significantly higher lesion counts were observed for coinfection cases than for VZV-alone but less than MPX-alone cases. Discernible differences in symptom and rash severity were detected for coinfection cases compared with those with MPX or VZV alone. Findings indicate infection with both MPX and VZV could modulate infection severity. Collection of multiple lesion samples allows for the opportunity to detect coinfections. As this program continues, it will be important to continue these procedures to assess variations in the proportion of coinfected cases over time.
Collapse
Affiliation(s)
- Christine M Hughes
- 1Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Lindy Liu
- 2Bacterial Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia.,3Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Whitni B Davidson
- 1Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Kay W Radford
- 4Viral Vaccine Preventable Diseases Branch, Division of Viral Diseases, National Center for Immunizations and Respiratory Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Kimberly Wilkins
- 1Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Benjamin Monroe
- 1Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Maureen G Metcalfe
- 3Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Toutou Likafi
- 5Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo
| | | | - Joelle Kabamba
- 7U.S. Centers for Disease Control and Prevention, Kinshasa, Democratic Republic of Congo
| | - Beatrice Nguete
- 5Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo
| | - Jean Malekani
- 8Department of Biology, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Elisabeth Pukuta
- 9Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | - Stomy Karhemere
- 9Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | | | | | - Mary G Reynolds
- 1Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - D Scott Schmid
- 4Viral Vaccine Preventable Diseases Branch, Division of Viral Diseases, National Center for Immunizations and Respiratory Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Andrea M McCollum
- 1Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
6
|
Hughes CM, Liu L, Davidson WB, Radford KW, Wilkins K, Monroe B, Metcalfe MG, Likafi T, Lushima RS, Kabamba J, Nguete B, Malekani J, Pukuta E, Karhemere S, Muyembe Tamfum JJ, Okitolonda Wemakoy E, Reynolds MG, Schmid DS, McCollum AM. A Tale of Two Viruses: Coinfections of Monkeypox and Varicella Zoster Virus in the Democratic Republic of Congo. Am J Trop Med Hyg 2020; 104:604-611. [PMID: 33289470 PMCID: PMC7866336 DOI: 10.4269/ajtmh.20-0589] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/17/2020] [Indexed: 12/16/2022] Open
Abstract
Recent enhanced monkeypox (MPX) surveillance in the Democratic Republic of Congo, where MPX is endemic, has uncovered multiple cases of MPX and varicella zoster virus (VZV) coinfections. The purpose of this study was to verify if coinfections occur and to characterize the clinical nature of these cases. Clinical, epidemiological, and laboratory results were used to investigate MPX/VZV coinfections. A coinfection was defined as a patient with at least one Orthopoxvirus/MPX-positive sample and at least one VZV-positive sample within the same disease event. Between September 2009 and April 2014, 134 of the 1,107 (12.1%) suspected MPX cases were confirmed as MPX/VZV coinfections. Coinfections were more likely to report symptoms than VZV-alone cases and less likely than MPX-alone cases. Significantly higher lesion counts were observed for coinfection cases than for VZV-alone but less than MPX-alone cases. Discernible differences in symptom and rash severity were detected for coinfection cases compared with those with MPX or VZV alone. Findings indicate infection with both MPX and VZV could modulate infection severity. Collection of multiple lesion samples allows for the opportunity to detect coinfections. As this program continues, it will be important to continue these procedures to assess variations in the proportion of coinfected cases over time.
Collapse
Affiliation(s)
- Christine M. Hughes
- Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Lindy Liu
- Bacterial Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Whitni B. Davidson
- Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Kay W. Radford
- Viral Vaccine Preventable Diseases Branch, Division of Viral Diseases, National Center for Immunizations and Respiratory Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Kimberly Wilkins
- Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Benjamin Monroe
- Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Maureen G. Metcalfe
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Toutou Likafi
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo
| | | | - Joelle Kabamba
- U.S. Centers for Disease Control and Prevention, Kinshasa, Democratic Republic of Congo
| | - Beatrice Nguete
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo
| | - Jean Malekani
- Department of Biology, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Elisabeth Pukuta
- Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | - Stomy Karhemere
- Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | | | | | - Mary G. Reynolds
- Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - D. Scott Schmid
- Viral Vaccine Preventable Diseases Branch, Division of Viral Diseases, National Center for Immunizations and Respiratory Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Andrea M. McCollum
- Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
7
|
Ramachandran V, Elliott SC, Rogers KL, Cohrs RJ, Weinberger M, Jackson W, Carpenter JE, Grose C, Bonthius DJ. Varicella Vaccine Meningitis as a Complication of Herpes Zoster in Twice-Immunized Immunocompetent Adolescents. J Child Neurol 2020; 35:889-895. [PMID: 32677551 PMCID: PMC7549284 DOI: 10.1177/0883073820938597] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/12/2020] [Accepted: 06/03/2020] [Indexed: 12/20/2022]
Abstract
Varicella-zoster virus vaccination is recommended for virtually all young children in the United States, Canada, and several other countries. Varicella vaccine is a live attenuated virus that retains some of its neurotropic properties. Herpes zoster caused by vaccine virus still occurs in immunized children, although the rate is much lower than in children who had wild-type varicella. It was commonly thought that 2 varicella vaccinations would protect children against the most serious complication of meningitis following herpes zoster; however, 2 meningitis cases have already been published. We now report a third case of varicella vaccine meningitis and define risk factors shared by all 3 immunized adolescents. The diagnosis in cerebrospinal fluid in this third case was verified by amplifying and sequencing portions of the viral genome, to document fixed alleles found only in the vaccine strain. Viral antibody was also detected in the cerebrospinal fluid by confocal microscopy. When compared with the other 2 cases, remarkably all 3 were 14 years old when meningitis occurred. All 3 were treated with intravenous acyclovir, with complete recovery. The adolescent in our case report also had recurrent asthma, which was treated with both prednisone tablets and beclomethasone inhaler before onset of meningitis. When the 3 cases were considered together, they suggested that immunity to varicella-zoster virus may be waning sufficiently in some twice-immunized adolescents to make them vulnerable to varicella vaccine virus reactivation and subsequent meningitis. This complication rarely happens in children after wild-type varicella.
Collapse
Affiliation(s)
- Veena Ramachandran
- Division of Infectious Diseases, Blank Children’s Hospital, Des Moines, IA, USA
| | - Stephen C. Elliott
- Division of Hematology-Oncology, Blank Children’s Hospital, Des Moines, IA, USA
| | - Kathie L. Rogers
- Clinical Microbiology Laboratory, Blank Children’s Hospital, Des Moines, IA, USA
| | - Randall J. Cohrs
- Department of Neurology, University of Colorado Anschutz Medical
Campus, Aurora, CO, USA
| | - Miles Weinberger
- Division of Pulmonary Diseases, University of Iowa Children’s
Hospital, University of Iowa, Iowa City, IA, USA
| | - Wallen Jackson
- Division of Infectious Diseases/Virology, University of Iowa
Children’s Hospital, University of Iowa, Iowa City, IA, USA
| | - John E. Carpenter
- Division of Infectious Diseases/Virology, University of Iowa
Children’s Hospital, University of Iowa, Iowa City, IA, USA
| | - Charles Grose
- Division of Infectious Diseases/Virology, University of Iowa
Children’s Hospital, University of Iowa, Iowa City, IA, USA
| | - Daniel J. Bonthius
- Division of Child Neurology, University of Iowa Children’s Hospital, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
8
|
Harpaz R. Teach your parents well: Pediatric recipients of varicella vaccines yield insights for adults regarding herpes zoster. Vaccine 2020; 38:5877-5879. [DOI: 10.1016/j.vaccine.2020.07.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 11/28/2022]
|
9
|
Jensen NJ, Depledge DP, Ng TFF, Leung J, Quinlivan M, Radford KW, Folster J, Tseng HF, LaRussa P, Jacobsen SJ, Breuer J, Schmid DS. Analysis of the reiteration regions (R1 to R5) of varicella-zoster virus. Virology 2020; 546:38-50. [PMID: 32452416 DOI: 10.1016/j.virol.2020.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/12/2020] [Accepted: 03/31/2020] [Indexed: 10/24/2022]
Abstract
The varicella-zoster virus (VZV) genome, comprises both unique and repeated regions. The genome also includes reiteration regions, designated R1 to R5, which are tandemly repeating sequences termed elements. These regions represent an understudied feature of the VZV genome. The R4 region is duplicated, with one copy in the internal repeat short (IRs) which we designated R4A and a second copy in the terminal repeat short (TRs) termed R4B. We developed primers to amplify and Sanger sequence these regions, including independent amplification of both R4 regions. Reiteration regions from >80 cases of PCR-confirmed shingles were sequenced and analyzed. Complete genome sequences for the remaining portions of these viruses were determined using Illumina MiSeq. We identified 28 elements not previously reported, including at least one element for each R region. Length heterogeneity was substantial in R3, R4A and R4B. Length heterogeneity between the two copies of R4 was common.
Collapse
Affiliation(s)
- Nancy J Jensen
- Division of Viral Diseases, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Daniel P Depledge
- Division of Infection and Immunity, University College London, London, United Kingdom; Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Terry Fei Fan Ng
- Division of Viral Diseases, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jessica Leung
- Division of Viral Diseases, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Mark Quinlivan
- Division of Viral Diseases, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Kay W Radford
- Division of Viral Diseases, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jennifer Folster
- Division of Viral Diseases, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Hung-Fu Tseng
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Philip LaRussa
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Steven J Jacobsen
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Judith Breuer
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - D Scott Schmid
- Division of Viral Diseases, National Center for Immunizations and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
10
|
Rosamilia LL. Herpes Zoster Presentation, Management, and Prevention: A Modern Case-Based Review. Am J Clin Dermatol 2020; 21:97-107. [PMID: 31741185 DOI: 10.1007/s40257-019-00483-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Herpes zoster (HZ) is a common cutaneous entity with protean clinical presentations, management options, complication rates, and prevention strategies, all of which are rife with dogma. During an inpatient consultation for HZ, have you ever been approached by a frantic staff or family member, worried that a pregnant, elderly, or infant contact will be 'infected' if they get too close? Have your patients ever asked you about their risk of having HZ twice, or claimed that they have frequent 'recurrences'? In what timeline should antiviral therapy be employed? Is there evidence for prednisone or gabapentin in acute HZ treatment? Who should be vaccinated against HZ and what are the benefits and risks? In case-based form, these and other complex but common scenarios will be examined using clinical and viral mechanistic clues, along with updated treatment and prevention guidelines, to provide a modern HZ case management compendium, comprehensive of the diverse age and health populations now presenting with this condition.
Collapse
|
11
|
Harbecke R, Jensen NJ, Depledge DP, Johnson GR, Ashbaugh ME, Schmid DS, Breuer J, Levin MJ, Oxman MN. Recurrent herpes zoster in the Shingles Prevention Study: Are second episodes caused by the same varicella-zoster virus strain? Vaccine 2020; 38:150-157. [PMID: 31679866 DOI: 10.1016/j.vaccine.2019.10.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/01/2019] [Accepted: 10/15/2019] [Indexed: 11/17/2022]
Abstract
Herpes zoster (HZ) is caused by reactivation of varicella zoster virus (VZV) that established latency in sensory and autonomic neurons during primary infection. In the Shingles Prevention Study (SPS), a large efficacy trial of live attenuated Oka/Merck zoster vaccine (ZVL), PCR-confirmed second episodes of HZ occurred in two of 660 placebo and one of 321 ZVL recipients with documented HZ during a mean follow-up of 3.13 years. An additional two ZVL recipients experienced a second episode of HZ in the Long-Term Persistence Substudy. All episodes of HZ were caused by wild-type VZV. The first and second episodes of HZ occurred in different dermatomes in each of these five participants, with contralateral recurrences in two. Time between first and second episodes ranged from 12 to 28 months. One of the five participants, who was immunocompetent on study enrollment, was immunocompromised at the onset of his first and second episodes of HZ. VZV DNA isolated from rash lesions from the first and second episodes of HZ was used to sequence the full-length VZV genomes. For the unique-sequence regions of the VZV genome, we employed target enrichment of VZV DNA, followed by deep sequencing. For the reiteration regions, we used PCR amplification and Sanger sequencing. Our analysis and comparison of the VZV genomes from the first and second episodes of HZ in each of the five participants indicate that both episodes were caused by the same VZV strain. This is consistent with the extraordinary stability of VZV during the replication phase of varicella and the subsequent establishment of latency in sensory ganglia throughout the body. Our observations also indicate that VZV is stable during the persistence of latency and the subsequent reactivation and replication that results in HZ.
Collapse
Affiliation(s)
- Ruth Harbecke
- Department of Veterans Affairs (VA) San Diego Healthcare System, San Diego, CA, USA; Department of Medicine, University of California San Diego, San Diego, CA, USA.
| | - Nancy J Jensen
- Centers for Disease Control and Prevention, Division of Viral Diseases, Atlanta, GA, USA
| | - Daniel P Depledge
- Division of Infection and Immunity, University College London, London, UK; Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - Gary R Johnson
- Cooperative Studies Program Coordinating Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Mark E Ashbaugh
- Department of Veterans Affairs (VA) San Diego Healthcare System, San Diego, CA, USA
| | - D Scott Schmid
- Centers for Disease Control and Prevention, Division of Viral Diseases, Atlanta, GA, USA
| | - Judith Breuer
- Division of Infection and Immunity, University College London, London, UK
| | - Myron J Levin
- Department of Medicine and Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Michael N Oxman
- Department of Veterans Affairs (VA) San Diego Healthcare System, San Diego, CA, USA; Department of Medicine, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
12
|
Egawa G, Egawa K, Kabashima K. Case of chickenpox in which varicella zoster virus genotype E was identified for the first time in Japan. J Dermatol 2019; 47:54-57. [PMID: 31742720 DOI: 10.1111/1346-8138.15153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/23/2019] [Indexed: 02/05/2023]
Abstract
Varicella zoster virus (VZV) is now classified into seven genotypes, and type J (clade 2) is known as an exclusively prevalent genotype in Japan. Here, we describe an adult Japanese patient who was suffering from chickenpox caused by VZV type E, the most prevalent genotype in Western Europe. Because the eruptions were distributed over the trunk and limbs and the patient had a high titer of immunoglobulin G against VZV, we diagnosed this case as secondary VZV infection. To investigate the current prevalence of VZV genotypes in Japan, we examined the genotype of VZV in an additional 49 Japanese varicella/zoster patients who visited our hospital during 2018-2019. We found that VZV type E was still an exceptionally rare genotype (1/50) in Japan. Because foreign nationals living in Japan who carry VZV genotypes other than type J are increasing in number, secondary chickenpox may increase in Japan in the near future, as well as in the USA where multiple VZV genotypes are distributed.
Collapse
Affiliation(s)
- Gyohei Egawa
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kiyofumi Egawa
- Amakusa Dermatology and Internal Medicine Clinic, Kamiamakusa, Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
13
|
Harpaz R. Do varicella vaccination programs change the epidemiology of herpes zoster? A comprehensive review, with focus on the United States. Expert Rev Vaccines 2019; 18:793-811. [PMID: 31318605 DOI: 10.1080/14760584.2019.1646129] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Introduction: Policy-makers in many countries have been wary of introducing varicella vaccination programs because of concerns that reduced exposures to varicella-zoster virus could increase herpes zoster (HZ) incidence. The U.S. introduced varicella vaccination in 1996 and has empiric evidence regarding this concern. Areas covered: This comprehensive review provides background emphasizing the epidemiology of varicella and of HZ in the U.S. before and after the introduction of their respective vaccines. The epidemiology is complex, and interpretation is complicated by methodologic challenges, by unexplained increases in age-specific HZ incidence that preceded varicella vaccination, and by introduction of vaccines for prevention of HZ. Nonetheless, observations from studies using different platforms and designs have yielded consistent findings, suggesting they are robust. Expert opinion: There has been no evidence that the U.S. varicella vaccination program increased HZ incidence in the general adult population over baseline trends. Furthermore, HZ incidence in children is declining. The U.S. experience can inform the development of new generations of models to predict HZ trends. More importantly, it provides reassurance for countries considering varicella vaccination that an effective program can reduce varicella morbidity and mortality while reducing the likelihood of HZ among children, and potentially, over time, across the entire population.
Collapse
Affiliation(s)
- Rafael Harpaz
- a Division of Viral Diseases, Centers for Disease Control and Prevention , Atlanta , GA , USA
| |
Collapse
|
14
|
Ashi A, Ali A, Alzahrani M, Ali J, Albar R. Herpes Zoster Eruption in an Otherwise Healthy Child: A Case Report. Cureus 2019; 11:e5194. [PMID: 31463164 PMCID: PMC6701888 DOI: 10.7759/cureus.5194] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Varicella-zoster virus (VZV; human herpesvirus 3) is a herpesvirus that causes infection in humans. The reactivation of latent VZV manifests as herpes zoster or shingles. In immunocompetent children, reactivation is rare, as increasing age is the most common risk factor for reactivation. The complications of infection include post-herpetic neuralgia and neurological sequelae as well as a bacterial infection of the crusted skin. We present a case of an otherwise healthy 13-year-old child with herpes zoster and multiple risk factors, who was managed successfully, in order to expand the limited literature. The patient presented with a painful vesicular rash, which appeared as multiple grouped vesicles on an erythematous base spreading over the right half of the face. The diagnosis of herpetic (varicella) vesiculobullous dermatitis was confirmed by biopsy and the patient was started on acyclovir and clindamycin with gradual improvement and resolution of his symptoms. Reactivation of VZV is considered a consequence of decreased cell-mediated immunity. However, the reason for reactivation in immunocompetent children remains unclear. In the present case, the patient may have become exposed in utero, through vaccination, or as a result of major facial trauma sustained at the age of six years.
Collapse
Affiliation(s)
- Abdullah Ashi
- Pediatrics, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Jeddah, SAU
| | - Abdullah Ali
- Pediatrics, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Jeddah, SAU
| | - Mohammed Alzahrani
- Pediatrics, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Jeddah, SAU
| | - Jumanah Ali
- Pediatrics, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Jeddah, SAU
| | - Rawia Albar
- Pediatrics, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Jeddah, SAU
| |
Collapse
|
15
|
Levin MJ, Cai GY, Lee KS, Rouphael NG, Mehta AK, Canniff J, Mulligan MJ, Weinberg A. Varicella-Zoster Virus DNA in Blood After Administration of Herpes Zoster Vaccine. J Infect Dis 2019; 217:1055-1059. [PMID: 29409017 DOI: 10.1093/infdis/jix653] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/16/2017] [Indexed: 01/20/2023] Open
Abstract
We studied the relationship between varicella-zoster virus (VZV) DNAemia and development of VZV-specific immunity after administration of live-attenuated zoster vaccine. VZV-DNAemia, detected by polymerase chain reaction (PCR), and VZV-specific effector (Teff) and memory (Tmem) T cells, was measured in 67 vaccinees. PCR was positive in 56% (9 direct, 28 nested) on day 1 and in 16% (1 direct, 10 nested) on day 14. Teff progressively increased in direct-PCR-positive vaccinees up to day 30, but Tmem did not. Conversely, Tmem, but not Teff, increased in direct-PCR-negative vaccinees on day 7. The kinetics of these immune responses and VZV DNAemia suggested that direct-PCR sample positive represented viremia.
Collapse
Affiliation(s)
- Myron J Levin
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora.,Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora
| | - Guang-Yun Cai
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora
| | - Katherine S Lee
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora
| | - Nadine G Rouphael
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta and Hope Clinic, Emory Vaccine Center, Decatur, Georgia
| | - Aneesh K Mehta
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta and Hope Clinic, Emory Vaccine Center, Decatur, Georgia
| | - Jennifer Canniff
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora
| | - Mark J Mulligan
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta and Hope Clinic, Emory Vaccine Center, Decatur, Georgia
| | - Adriana Weinberg
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora.,Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora.,Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora
| |
Collapse
|
16
|
Leung J, McCollum AM, Radford K, Hughes C, Lopez AS, Guagliardo SAJ, Nguete B, Likafi T, Kabamba J, Malekani J, Shongo Lushima R, Pukuta E, Karhemere S, Muyembe Tamfum JJ, Reynolds MG, Wemakoy Okitolonda E, Schmid DS, Marin M. Varicella in Tshuapa Province, Democratic Republic of Congo, 2009-2014. Trop Med Int Health 2019; 24:839-848. [PMID: 31062445 PMCID: PMC8786670 DOI: 10.1111/tmi.13243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To describe varicella cases in Tshuapa Province of the Democratic Republic of the Congo identified during monkeypox surveillance. METHODS Demographic, clinical and epidemiological data were collected from each suspected monkeypox case 2009-2014. Samples were tested by PCR for both Orthopoxviruses and varicella-zoster virus (VZV); a subset of VZV-positive samples was genotyped. We defined a varicella case as a rash illness with laboratory-confirmed VZV. RESULTS There were 366 varicella cases were identified; 66% were ≤19 years old. Most patients had non-typical varicella rash with lesions reported as the same size and stage of evolution (86%), deep and profound (91%), on palms of hands and/or soles of feet (86%) and not itchy (49%). Many had non-typical signs and symptoms, such as lymphadenopathy (70%) and sensitivity to light (23%). A higher proportion of persons aged ≥20 years than persons aged ≤19 years had ≥50 lesions (79% vs. 65%, P = 0.007) and were bedridden (15% vs. 9%, P = 0.056). All VZV isolates genotyped from 79 varicella cases were clade 5. During the surveillance period, one possible VZV-related death occurred in a 7-year-old child. CONCLUSIONS A large proportion of patients presented with non-typical varicella rash and clinical signs and symptoms, highlighting challenges identifying varicella in an area with endemic monkeypox. Continued surveillance and laboratory diagnosis will help in rapid identification and control of both monkeypox and varicella and improve our understanding of varicella epidemiology in Africa.
Collapse
Affiliation(s)
- Jessica Leung
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention Atlanta GA USA
| | - Andrea M. McCollum
- National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta GA USA
| | - Kay Radford
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention Atlanta GA USA
| | - Christine Hughes
- National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta GA USA
| | - Adriana S Lopez
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention Atlanta GA USA
| | - Sarah Anne J. Guagliardo
- National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta GA USA
- Epidemic Intelligence Service, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA, 30333, United States
| | - Beatrice Nguete
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo
| | - Toutou Likafi
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo
| | - Joelle Kabamba
- U.S. Centers for Disease Control and Prevention, Kinshasa, Democratic Republic of Congo
| | - Jean Malekani
- University of Kinshasa, Department of Biology, Kinshasa, Democratic Republic of Congo
| | | | - Elisabeth Pukuta
- Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | - Stomy Karhemere
- Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | | | - Mary G. Reynolds
- National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta GA USA
| | | | - D Scott Schmid
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention Atlanta GA USA
| | - Mona Marin
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention Atlanta GA USA
| |
Collapse
|
17
|
Heath G, Depledge DP, Brown JR, Hale AD, Tutil H, Williams R, Breuer J. Acute Retinal Necrosis Caused by the Zoster Vaccine Virus. Clin Infect Dis 2018; 65:2122-2125. [PMID: 29020238 PMCID: PMC5849943 DOI: 10.1093/cid/cix683] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 09/04/2017] [Indexed: 12/14/2022] Open
Abstract
We report acute retinal necrosis caused by the vaccine Oka strain following immunization of a 78-year-old woman with live zoster vaccine. Whole genome sequencing confirmed the ocular vOka strain to be derived from the vaccine and excluded the presence of new mutations or recombination with wild-type Varicella zoster virus.
Collapse
Affiliation(s)
- Gregory Heath
- Medical Ophthalmology, York Teaching Hospital NHS Foundation Trust, York, England
| | | | - Julianne R Brown
- Microbiology, Virology and Infection Control (VZV Typing Laboratory), Great Ormond St Hospital, London, England
| | - Anthony D Hale
- Virology, Leeds Teaching Hospitals NHS Trust, Leeds, England
| | - Helena Tutil
- Infection and Immunity, University College London, England
| | | | - Judith Breuer
- Infection and Immunity, University College London, England.,Microbiology, Virology and Infection Control (VZV Typing Laboratory), Great Ormond St Hospital, London, England
| |
Collapse
|
18
|
Redelman-Sidi G, Michielin O, Cervera C, Ribi C, Aguado JM, Fernández-Ruiz M, Manuel O. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Immune checkpoint inhibitors, cell adhesion inhibitors, sphingosine-1-phosphate receptor modulators and proteasome inhibitors). Clin Microbiol Infect 2018; 24 Suppl 2:S95-S107. [PMID: 29427804 DOI: 10.1016/j.cmi.2018.01.030] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/18/2018] [Accepted: 01/27/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND The present review is part of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Infections in Compromised Hosts (ESGICH) consensus document on the safety of targeted and biological therapies. AIMS To review, from an infectious diseases perspective, the safety profile of immune checkpoint inhibitors, LFA-3-targeted agents, cell adhesion inhibitors, sphingosine-1-phosphate receptor modulators and proteasome inhibitors, and to suggest preventive recommendations. SOURCES Computer-based Medline searches with MeSH terms pertaining to each agent or therapeutic family. CONTENT T-lymphocyte-associated antigen 4 (CTLA-4) and programmed death (PD)-1/PD-1 ligand 1 (PD-L1)-targeted agents do not appear to intrinsically increase the risk of infection but can induce immune-related adverse effects requiring additional immunosuppression. Although CD4+ T-cell lymphopenia is associated with alefacept, no opportunistic infections have been observed. Progressive multifocal leukoencephalopathy (PML) may occur during therapy with natalizumab (anti-α4-integrin monoclonal antibody (mAb)) and efalizumab (anti-CD11a mAb), but no cases have been reported to date with vedolizumab (anti-α4β7 mAb). In patients at high risk for PML (positive anti-JC polyomavirus serology with serum antibody index >1.5 and duration of therapy ≥48 months), the benefit-risk ratio of continuing natalizumab should be carefully considered. Fingolimod induces profound peripheral blood lymphopenia and increases the risk of varicella zoster virus (VZV) infection. Prophylaxis with (val)acyclovir and VZV vaccination should be considered. Proteasome inhibitors also increase the risk of VZV infection, and antiviral prophylaxis with (val)acyclovir is recommended. Anti-Pneumocystis prophylaxis may be considered in myeloma multiple patients with additional risk factors (i.e. high-dose corticosteroids). IMPLICATIONS Clinicians should be aware of the risk of immune-related adverse effects and PML in patients receiving immune checkpoint and cell adhesion inhibitors respectively.
Collapse
Affiliation(s)
- G Redelman-Sidi
- Service of Infectious Disease, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA.
| | - O Michielin
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - C Cervera
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - C Ribi
- Department of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - J M Aguado
- Unit of Infectious Diseases, Hospital Universitario '12 de Octubre', Instituto de Investigación Hospital '12 de Octubre' (i+12), Madrid, Spain; Spanish Network for Research in Infectious Diseases (REIPI RD16/0016), Instituto de Salud Carlos III, Madrid, Spain
| | - M Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario '12 de Octubre', Instituto de Investigación Hospital '12 de Octubre' (i+12), Madrid, Spain; Spanish Network for Research in Infectious Diseases (REIPI RD16/0016), Instituto de Salud Carlos III, Madrid, Spain
| | - O Manuel
- Department of Infectious Diseases, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
19
|
Willis ED, Woodward M, Brown E, Popmihajlov Z, Saddier P, Annunziato PW, Halsey NA, Gershon AA. Herpes zoster vaccine live: A 10 year review of post-marketing safety experience. Vaccine 2017; 35:7231-7239. [PMID: 29174682 DOI: 10.1016/j.vaccine.2017.11.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/06/2017] [Accepted: 11/07/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Zoster vaccine is a single dose live, attenuated vaccine (ZVL) indicated for individuals ≥50 years-old for the prevention of herpes zoster (HZ). Safety data from clinical trials and post-licensure studies provided reassurance that ZVL is generally safe and well tolerated. The objective of this review was to provide worldwide post-marketing safety information following 10 years of use and >34 million doses distributed. METHODS All post-marketing adverse experience (AE) reports received worldwide between 02-May-2006 and 01-May-2016 from healthcare professionals following vaccination with ZVL and submitted to the MSD AE global safety database, were analyzed. RESULTS A total of 23,556 AE reports, 93% non-serious, were reported. Local injection site reactions (ISRs), with a median time-to-onset of 2 days, were the most frequently reported AEs followed by HZ. The majority of HZ reports were reported within 2 weeks of vaccination and considered, based on time-to-onset, pathogenesis of HZ, and data from clinical trials, to be caused by wild-type varicella-zoster virus (VZV). HZ confirmed by PCR analysis to be VZV Oka/Merck vaccine-strain was identified in an immunocompetent individual 8 months postvaccination and in 4 immunocompromised individuals. Disseminated HZ was reported very rarely (<1%) with 38% occurring in immunocompromised individuals. All reports of disseminated HZ confirmed by PCR as VZV Oka/Merck vaccine-strain were in individuals with immunosuppressive conditions and/or therapy at the time of vaccination. CONCLUSIONS The safety profile of ZVL, following 10 years of post-marketing use, was favorable and consistent with that observed in clinical trials and post-licensure studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Neal A Halsey
- Institute for Vaccine Safety, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Anne A Gershon
- Columbia University College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
20
|
Harada K, Heaton H, Chen J, Vazquez M, Meyer J. Zoster vaccine-associated primary varicella infection in an immunocompetent host. BMJ Case Rep 2017; 2017:bcr-2017-221166. [PMID: 28830902 DOI: 10.1136/bcr-2017-221166] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
A 64-year-old immunocompetent man developed a widespread pruritic and vesicular rash 2 weeks after receiving the zoster vaccine (Zostavax). He had fever, bandaemia with normal total white blood cell count and mild transaminitis. PCR testing of serum and skin was positive for varicella zoster virus (VZV), while serum VZV IgG was negative. The analysis of single nucleotide polymorphism by PCR and sequencing from the skin swab was consistent with the vaccine strain. The patient received 1 week of intravenous acyclovir and was discharged after all lesions had crusted. He continues to do well on follow-up with no significant complications.
Collapse
Affiliation(s)
- Kaoru Harada
- Department of Internal Medicine, Yale-New Haven Hospital, New Haven, Connecticut, USA
| | - Henry Heaton
- Department of Dermatology, Yale-New Haven Hospital, New Haven, Connecticut, USA
| | - Jason Chen
- Department of Pathology & Cell Biology, Columbia University College of Physicians & Surgeons, New York, New York, USA
| | - Marietta Vazquez
- Department of Pediatrics, Infectious Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jaimie Meyer
- Department of Medicine, Infectious Diseases, AIDS Program, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
21
|
Jensen NJ, Rivailler P, Tseng HF, Quinlivan ML, Radford K, Folster J, Harpaz R, LaRussa P, Jacobsen S, Scott Schmid D. Revisiting the genotyping scheme for varicella-zoster viruses based on whole-genome comparisons. J Gen Virol 2017; 98:1434-1438. [PMID: 28613146 DOI: 10.1099/jgv.0.000772] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We report whole-genome sequences (WGSs) for four varicella-zoster virus (VZV) samples from a shingles study conducted by Kaiser Permanente of Southern California. Comparative genomics and phylogenetic analysis of all published VZV WGSs revealed that strain KY037798 is in clade IX, which shall henceforth be designated clade 9. Previously published single nucleotide polymorphisms (SNP)-based genotyping schemes fail to discriminate between clades 6 and VIII and employ positions that are not clade-specific. We provide an updated list of clade-specific positions that supersedes the list determined at the 2008 VZV nomenclature meeting. Finally, we propose a new targeted genotyping scheme that will discriminate the circulating VZV clades with at least a twofold redundancy. Genotyping strategies using a limited set of targeted SNPs will continue to provide an efficient 'first pass' method for VZV strain surveillance as vaccination programmes for varicella and zoster influence the dynamics of VZV transmission.
Collapse
Affiliation(s)
- Nancy J Jensen
- Centers for Disease Control and Prevention, National Center for Immunizations and Respiratory Diseases, Division of Viral Diseases, Atlanta, GA, USA
| | - Pierre Rivailler
- Centers for Disease Control and Prevention, National Center for Immunizations and Respiratory Diseases, Division of Viral Diseases, Atlanta, GA, USA
| | - Hung Fu Tseng
- Kaiser Permanente of Southern California, Department of Research and Evaluation, Pasadena, CA, USA
| | - Mark L Quinlivan
- Centers for Disease Control and Prevention, National Center for Immunizations and Respiratory Diseases, Division of Viral Diseases, Atlanta, GA, USA.,Present address: Labtech International Ltd, Uckfield, East Sussex, UK
| | - Kay Radford
- Centers for Disease Control and Prevention, National Center for Immunizations and Respiratory Diseases, Division of Viral Diseases, Atlanta, GA, USA
| | - Jennifer Folster
- Centers for Disease Control and Prevention, National Center for Immunizations and Respiratory Diseases, Division of Viral Diseases, Atlanta, GA, USA
| | - Rafael Harpaz
- Centers for Disease Control and Prevention, National Center for Immunizations and Respiratory Diseases, Division of Viral Diseases, Atlanta, GA, USA
| | - Philip LaRussa
- Department of Pediatrics, Columbia University, College of Physicians and Surgeons,, New York, NY, USA
| | - Steven Jacobsen
- Kaiser Permanente of Southern California, Department of Research and Evaluation, Pasadena, CA, USA
| | - D Scott Schmid
- Centers for Disease Control and Prevention, National Center for Immunizations and Respiratory Diseases, Division of Viral Diseases, Atlanta, GA, USA
| |
Collapse
|
22
|
ORF7 of Varicella-Zoster Virus Is Required for Viral Cytoplasmic Envelopment in Differentiated Neuronal Cells. J Virol 2017; 91:JVI.00127-17. [PMID: 28356523 DOI: 10.1128/jvi.00127-17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/24/2017] [Indexed: 12/20/2022] Open
Abstract
Although a varicella-zoster virus (VZV) vaccine has been used for many years, the neuropathy caused by VZV infection is still a major health concern. Open reading frame 7 (ORF7) of VZV has been recognized as a neurotropic gene in vivo, but its neurovirulent role remains unclear. In the present study, we investigated the effect of ORF7 deletion on VZV replication cycle at virus entry, genome replication, gene expression, capsid assembly and cytoplasmic envelopment, and transcellular transmission in differentiated neural progenitor cells (dNPCs) and neuroblastoma SH-SY5Y (dSY5Y) cells. Our results demonstrate that the ORF7 protein is a component of the tegument layer of VZV virions. Deleting ORF7 did not affect viral entry, viral genome replication, or the expression of typical viral genes but clearly impacted cytoplasmic envelopment of VZV capsids, resulting in a dramatic increase of envelope-defective particles and a decrease in intact virions. The defect was more severe in differentiated neuronal cells of dNPCs and dSY5Y. ORF7 deletion also impaired transmission of ORF7-deficient virus among the neuronal cells. These results indicate that ORF7 is required for cytoplasmic envelopment of VZV capsids, virus transmission among neuronal cells, and probably the neuropathy induced by VZV infection.IMPORTANCE The neurological damage caused by varicella-zoster virus (VZV) reactivation is commonly manifested as clinical problems. Thus, identifying viral neurovirulent genes and characterizing their functions are important for relieving VZV related neurological complications. ORF7 has been previously identified as a potential neurotropic gene, but its involvement in VZV replication is unclear. In this study, we found that ORF7 is required for VZV cytoplasmic envelopment in differentiated neuronal cells, and the envelopment deficiency caused by ORF7 deletion results in poor dissemination of VZV among neuronal cells. These findings imply that ORF7 plays a role in neuropathy, highlighting a potential strategy to develop a neurovirulence-attenuated vaccine against chickenpox and herpes zoster and providing a new target for intervention of neuropathy induced by VZV.
Collapse
|
23
|
Herpes zoster: Risk and prevention during immunomodulating therapy. Joint Bone Spine 2017; 84:21-27. [DOI: 10.1016/j.jbspin.2016.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2016] [Indexed: 12/30/2022]
|
24
|
Jeon JS, Won YH, Kim IK, Ahn JH, Shin OS, Kim JH, Lee CH. Analysis of single nucleotide polymorphism among Varicella-Zoster Virus and identification of vaccine-specific sites. Virology 2016; 496:277-286. [PMID: 27376245 DOI: 10.1016/j.virol.2016.06.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/11/2016] [Accepted: 06/21/2016] [Indexed: 12/30/2022]
Abstract
Varicella-zoster virus (VZV) is a causative agent for chickenpox and zoster. Live attenuated vaccines have been developed based on Oka and MAV/06 strains. In order to understand the molecular mechanisms of attenuation, complete genome sequences of vaccine and wild-type strains were compared and single nucleotide polymorphism (SNP) was analyzed. ORF22 and ORF62 contained the highest number of SNPs. The detailed analysis of the SNPs suggested 24 potential vaccine-specific sites. All the mutational events found in vaccine-specific sites were transitional, and most of them were substitution of AT to GC pair. Interestingly, 18 of the vaccine-specific sites of the vaccine strains appeared to be genetically heterogeneous. The probability of a single genome of vaccine strain to contain all 24 vaccine-type sequences was calculated to be less than 4%. The average codon adaptation index (CAI) value of the vaccine strains was significantly lower than the CAI value of the clinical strains.
Collapse
Affiliation(s)
- Jeong Seon Jeon
- Department of Microbiology, Chungbuk National University, Cheongju, South Korea
| | - Youn Hee Won
- Department of Microbiology, Chungbuk National University, Cheongju, South Korea
| | - In Kyo Kim
- Department of Microbiology, Chungbuk National University, Cheongju, South Korea
| | - Jin Hyun Ahn
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Ok Sarah Shin
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Jung Hwan Kim
- Mogam Biotechnology Research Institute, Yongin, South Korea
| | - Chan Hee Lee
- Department of Microbiology, Chungbuk National University, Cheongju, South Korea.
| |
Collapse
|
25
|
Gershon AA, Gershon M. Varicella Zoster Virus and Giant Cell Arteritis. J Infect Dis 2016; 213:1859-61. [PMID: 27037085 DOI: 10.1093/infdis/jiw110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 03/15/2016] [Indexed: 12/30/2022] Open
Affiliation(s)
| | - Michael Gershon
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York
| |
Collapse
|
26
|
Ganacias K, Spriet S, Banks TA. Brief Communication: A 61-year-old Woman with Vesicular Eruption after Varicella Zoster Vaccination. ALLERGY & RHINOLOGY 2016; 7:96-8. [PMID: 27349562 PMCID: PMC5010440 DOI: 10.2500/ar.2016.7.0159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background Vesicular rashes are associated with a variety of infectious and noninfectious causes. Objective To discuss the differential diagnoses of vesicular rashes. Methods We present the clinical case of an adult woman who was immunocompetent and who developed several clear fluid-filled vesicles on her upper extremity within days of receiving the varicella zoster vaccine. Over the next several days, the skin eruption generalized, and she developed new lesions in various stages of healing. Results After a detailed history and further studies were obtained, a final diagnosis was made. Conclusion In patients who have recently been vaccinated, a high index of suspicion for an adverse vaccine reaction should be maintained.
Collapse
Affiliation(s)
- Karen Ganacias
- Department of Defense and Walter Reed National Military Medical Center System, Bethesda, Maryland
| | - Sarah Spriet
- Department of Defense and Walter Reed National Military Medical Center System, Bethesda, Maryland
| | - Taylor A. Banks
- Department of Defense and Walter Reed National Military Medical Center System, Bethesda, Maryland
| |
Collapse
|
27
|
Shafran SD. Live attenuated herpes zoster vaccine for HIV-infected adults. HIV Med 2015; 17:305-10. [PMID: 26315285 DOI: 10.1111/hiv.12311] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2015] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Multiple guidelines exist for the use of live viral vaccines for measles-mumps-rubella (MMR), varicella and yellow fever in people with HIV infections, but these guidelines do not make recommendations regarding live attenuated herpes zoster vaccine (LAHZV), which is approved for people over 50 years in the general population. LAHZV is made with the same virus used in varicella vaccine. The incidence of herpes zoster remains increased in people with HIV infection, even when on suppressive antiretroviral therapy, and a growing proportion of HIV-infected patients are over 50 years of age. The purpose of this article is to review the use of varicella vaccine and LAHZV in people with HIV infection and to make recommendations about the use of LAHZV in adults with HIV infection. METHODS A PubMed search was undertaken using the terms 'herpes zoster AND HIV' and 'varicella AND HIV'. Reference lists were also reviewed for pertinent citations. RESULTS Varicella vaccine is recommended in varicella-susceptible adults, as long as they have a CD4 count > 200 cells/μL, the same CD4 threshold used for MMR and yellow fever vaccines. No transmission of vaccine strain Varicella zoster virus has been documented in people with HIV infections with a CD4 count above this threshold. LAHZV was administered to 295 HIV-infected adults with a CD4 count > 200 cells/μL, and was safe and immunogenic with no cases of vaccine strain infection. CONCLUSIONS It is recommended that LAHZV be administered to HIV-infected adults with a CD4 count above 200 cells/μL, the same CD4 threshold used for other live attenuated viral vaccines.
Collapse
Affiliation(s)
- S D Shafran
- Division of Infectious Diseases, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
28
|
Cheetham TC, Marcy SM, Tseng HF, Sy LS, Liu ILA, Bixler F, Baxter R, Donahue JG, Naleway AL, Jacobsen SJ. Risk of Herpes Zoster and Disseminated Varicella Zoster in Patients Taking Immunosuppressant Drugs at the Time of Zoster Vaccination. Mayo Clin Proc 2015; 90:865-73. [PMID: 26051268 DOI: 10.1016/j.mayocp.2015.04.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/29/2015] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To determine the risks associated with zoster vaccine when administered to patients taking immunosuppressant medications. PATIENTS AND METHODS Patients enrolled in 1 of 7 managed care organizations affiliated with the Vaccine Safety Datalink between January 1, 2006, and December 31, 2009, were eligible. The exposure of interest was zoster vaccination in patients with current or remote immunosuppressant drug use. The primary outcomes were disseminated varicella zoster virus (VZV) and herpes zoster in the 42 days after vaccination. Automated data were collected on immunosuppressant drugs and baseline medical conditions. A logistic regression model using inverse probability treatment weights was used to estimate the odds of developing VZV or herpes zoster. RESULTS A total of 14,554 individuals had an immunosuppressant medication dispensed around the time of vaccination, including 4826 with current use and 9728 with remote use. Most patients were taking low-dose corticosteroids. No cases of disseminated VZV were found in the current or remote users. The risk of herpes zoster was elevated in the 42 days after vaccination in current vs remote users (adjusted odds ratio, 2.99; 95% CI, 1.58-5.70). CONCLUSION We found that patients taking immunosuppressant medications at the time of vaccination had a modest increased risk of herpes zoster in the 42 days after vaccination. The development of herpes zoster within 42 days after vaccination suggests that this is more likely due to reactivation of latent zoster virus than dissemination of the vaccine-derived varicella virus. These findings support the current zoster vaccination guidelines.
Collapse
Affiliation(s)
- T Craig Cheetham
- Research and Evaluation, Kaiser Permanente Southern California, Pasadena.
| | - S Michael Marcy
- Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - Hung-Fu Tseng
- Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - Lina S Sy
- Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - In-Lu Amy Liu
- Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - Felicia Bixler
- Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - Roger Baxter
- Division of Research, Kaiser Permanente Northern California, Oakland
| | | | | | - Steven J Jacobsen
- Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| |
Collapse
|
29
|
GABUTTI G, VALENTE N, SULCAJ N, STEFANATI A. Evaluation of efficacy and effectiveness of live attenuated zoster vaccine. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2014; 55:130-6. [PMID: 26137786 PMCID: PMC4718315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Herpes zoster (HZ) is a viral disease characterized by a dermatologic and neurologic involvement caused by the reactivation of the latent varicella zoster virus (VZV) acquired during primary infection (varicella). HZ incidence increases with age and is related to waning specific cell-mediated immunity (CMI). The most frequent complication of HZ is post-herpetic neuralgia (PHN) characterized by chronic pain lasting at least 30 days, with impact on patients' quality of life. Available treatments are quite unsatisfactory in reducing pain and length of the disease. The evaluation of the epidemiology, the debilitating complications (PHN), the suboptimal available treatments and the costs related to the diagnosis and clinical/therapeutic management of HZ patients have been the rationale for the search of an adequate preventive measure against this disease. The target of this intervention is to reduce the frequency and severity of HZ and related complications by stimulating CMI. Prevention has recently become possible with the live attenuated vaccine Oka/Merck, with an antigen content at least 10-fold higher than the antigen content of pediatric varicella vaccines. Clinical studies show a good level of efficacy and effectiveness, particularly against the burden of illness and PHN in all age classes. Accordingly to the summary of the characteristics of the product the zoster vaccine is indicated for the prevention of HZ and PHN in individuals 50 years of age or older and is effective and safe in subjects with a positive history of HZ.
Collapse
Affiliation(s)
- G. GABUTTI
- Correspondence: Giovanni Gabutti, Dept. of Medical Sciences, University of Ferrara, via Fossato di Mortara 64b, 44121 Ferrara, Italy - Tel. +39 0532 455568 - Fax +39 0532 205066 - E-mail:
| | | | | | | |
Collapse
|
30
|
Abortive intrabronchial infection of rhesus macaques with varicella-zoster virus provides partial protection against simian varicella virus challenge. J Virol 2014; 89:1781-93. [PMID: 25410871 DOI: 10.1128/jvi.03124-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
UNLABELLED Varicella-zoster virus (VZV) is a human neurotropic alphaherpesvirus and the etiological agent of varicella (chickenpox) and herpes zoster (HZ, shingles). Previously, inoculation of monkeys via the subcutaneous, intratracheal, intravenous, or oral-nasal-conjunctival routes did not recapitulate all the hallmarks of VZV infection, including varicella, immunity, latency, and reactivation. Intrabronchial inoculation of rhesus macaques (RMs) with simian varicella virus (SVV), a homolog of VZV, recapitulates virologic and immunologic hallmarks of VZV infection in humans. Given that VZV is acquired primarily via the respiratory route, we investigated whether intrabronchial inoculation of RMs with VZV would result in a robust model. Despite the lack of varicella and viral replication in either the lungs or whole blood, all four RMs generated an immune response characterized by the generation of VZV-specific antibodies and T cells. Two of 4 VZV-inoculated RMs were challenged with SVV to determine cross-protection. VZV-immune RMs displayed no varicella rash and had lower SVV viral loads and earlier and stronger humoral and cellular immune responses than controls. In contrast to the results for SVV DNA, no VZV DNA was detected in sensory ganglia at necropsy. In summary, following an abortive VZV infection, RMs developed an adaptive immune response that conferred partial protection against SVV challenge. These data suggest that a replication-incompetent VZV vaccine that does not establish latency may provide sufficient protection against VZV disease and that VZV vaccination of RMs followed by SVV challenge provides a model to evaluate new vaccines and therapeutics against VZV. IMPORTANCE Although VZV vaccine strain Oka is attenuated, it can cause mild varicella, establish latency, and in rare cases, reactivate to cause herpes zoster (HZ). Moreover, studies suggest that the HZ vaccine (Zostavax) only confers short-lived immunity. The development of more efficacious vaccines would be facilitated by a robust animal model of VZV infection. The data presented in this report show that intrabronchial inoculation of rhesus macaques (RMs) with VZV resulted in an abortive VZV infection. Nevertheless, all animals generated a humoral and cellular immune response that conferred partial cross-protection against simian varicella virus (SVV) challenge. Additionally, VZV DNA was not detected in the sensory ganglia, suggesting that viremia might be required for the establishment of latency. Therefore, VZV vaccination of RMs followed by SVV challenge is a model that will support the development of vaccines that boost protective T cell responses against VZV.
Collapse
|
31
|
Abstract
Shingles (or zoster) is a reactivation of an existing varicella-zoster virus (VZV) infection. During the initial infection, VZV causes a systemic disease known as varicella or chickenpox, and this initial infection normally occurs early in childhood in the absence of routine vaccination. Although varicella is normally a mild disease, shingles is associated with significant morbidity and some mortality, particularly in older people. The most significant severe consequence is post-herpetic neuralgia. There is an effective vaccine available for this, known as Zostavax, which is a live-attenuated VZV vaccine. Guidelines in the UK recommend that this is offered to everyone when they become 70 years of age, plus those aged 79 years as part of a catch-up campaign, with those between these ages not being eligible. It is important for all health-care professionals, including district and community nurses, to proactively promote this vaccine, so that those eligible can make an informed decision about whether to receive it.
Collapse
Affiliation(s)
- Edward Purssell
- Senior Lecturer, Florence Nightingale School of Nursing and Midwifery, King's College London
| |
Collapse
|
32
|
Tseng HF, Tartof S, Harpaz R, Luo Y, Sy LS, Hetcher RC, Jacobsen SJ. Vaccination against zoster remains effective in older adults who later undergo chemotherapy. Clin Infect Dis 2014; 59:913-9. [PMID: 25097079 DOI: 10.1093/cid/ciu498] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Approximately 40% of adults develop invasive cancer during their lifetimes, many of whom require chemotherapy. Herpes zoster (HZ) is common and often severe in patients undergoing chemotherapy, yet there are no data regarding whether these patients retain specific protection against HZ if they had previously received zoster vaccine. We conducted a study to determine whether zoster vaccine was effective in patients who subsequently underwent chemotherapy. METHODS The cohort study consisted of Kaiser Permanente Southern California members aged ≥60 years treated with chemotherapy. The exposure variable was receipt of zoster vaccine prior to initiation of chemotherapy. Incident HZ cases were identified using International Classification of Diseases, Ninth Revision diagnostic codes. HZ incidence rates were calculated; hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated using Cox proportional hazards regression models. RESULTS There were 91 and 583 HZ cases in the vaccinated and unvaccinated cohorts, respectively, yielding an incidence rate of 12.87 (95% CI, 10.48-15.80) vs 22.05 (95% CI, 20.33-23.92) per 1000 person-years. Thirty-month cumulative incidence was 3.28% in the vaccinated group and 5.34% in the unvaccinated group (P < .05). The adjusted HR for HZ was 0.58 (95% CI, .46-.73) and showed no significant variation by age, sex, or race. HZ incidence rates remained increased in the small subgroup of persons receiving zoster vaccine within 60 days before chemotherapy, but this was the only group affected by indication bias. No vaccinated patients underwent hospitalization for HZ, compared with 6 unvaccinated patients. CONCLUSIONS Zoster vaccine continues to protect against HZ if recipients later undergo chemotherapy. Our findings provide an additional rationale for offering zoster vaccine to indicated adults while they are immunocompetent.
Collapse
Affiliation(s)
- Hung Fu Tseng
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - Sara Tartof
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - Rafael Harpaz
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Yi Luo
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - Lina S Sy
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - Rulin C Hetcher
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - Steven J Jacobsen
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| |
Collapse
|
33
|
The varicella-zoster virus portal protein is essential for cleavage and packaging of viral DNA. J Virol 2014; 88:7973-86. [PMID: 24807720 DOI: 10.1128/jvi.00376-14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The varicella-zoster virus (VZV) open reading frame 54 (ORF54) gene encodes an 87-kDa monomer that oligomerizes to form the VZV portal protein, pORF54. pORF54 was hypothesized to perform a function similar to that of a previously described herpes simplex virus 1 (HSV-1) homolog, pUL6. pUL6 and the associated viral terminase are required for processing of concatemeric viral DNA and packaging of individual viral genomes into preformed capsids. In this report, we describe two VZV bacterial artificial chromosome (BAC) constructs with ORF54 gene deletions, Δ54L (full ORF deletion) and Δ54S (partial internal deletion). The full deletion of ORF54 likely disrupted essential adjacent genes (ORF53 and ORF55) and therefore could not be complemented on an ORF54-expressing cell line (ARPE54). In contrast, Δ54S was successfully propagated in ARPE54 cells but failed to replicate in parental, noncomplementing ARPE19 cells. Transmission electron microscopy confirmed the presence of only empty VZV capsids in Δ54S-infected ARPE19 cell nuclei. Similar to the HSV-1 genome, the VZV genome is composed of a unique long region (UL) and a unique short region (US) flanked by inverted repeats. DNA from cells infected with parental VZV (VZVLUC strain) contained the predicted UL and US termini, whereas cells infected with Δ54S contained neither. This result demonstrates that Δ54S is not able to process and package viral DNA, thus making pORF54 an excellent chemotherapeutic target. In addition, the utility of BAC constructs Δ54L and Δ54S as tools for the isolation of site-directed ORF54 mutants was demonstrated by recombineering single-nucleotide changes within ORF54 that conferred resistance to VZV-specific portal protein inhibitors. Importance: Antivirals with novel mechanisms of action would provide additional therapeutic options to treat human herpesvirus infections. Proteins involved in the herpesviral DNA encapsidation process have become promising antiviral targets. Previously, we described a series of N-α-methylbenzyl-N'-aryl thiourea analogs that target the VZV portal protein (pORF54) and prevent viral replication in vitro. To better understand the mechanism of action of these compounds, it is important to define the structural and functional characteristics of the VZV portal protein. In contrast to HSV, no VZV mutants have been described for any of the seven essential DNA encapsidation genes. The VZV ORF54 deletion mutant described in this study represents the first VZV encapsidation mutant reported to date. We demonstrate that the deletion mutant can serve as a platform for the isolation of portal mutants via recombineering and provide a strategy for more in-depth studies of VZV portal structure and function.
Collapse
|