1
|
Tommasi C, Yogev O, Yee MB, Drousioti A, Jones M, Ring A, Singh M, Dry I, Atkins O, Naeem AS, Kriplani N, Akbar AN, Haas JG, O'Toole EA, Kinchington PR, Breuer J. Upregulation of keratin 15 is required for varicella-zoster virus replication in keratinocytes and is attenuated in the live attenuated vOka vaccine strain. Virol J 2024; 21:253. [PMID: 39385182 PMCID: PMC11465976 DOI: 10.1186/s12985-024-02514-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 09/22/2024] [Indexed: 10/11/2024] Open
Abstract
Varicella-zoster virus (VZV) is the etiological agent of chickenpox and shingles, diseases characterised by epidermal virus replication in skin and mucosa and the formation of blisters. We have previously shown that VZV infection has a profound effect on keratinocyte differentiation, altering the normal pattern of epidermal gene expression. In particular, VZV infection reduces expression of suprabasal keratins 1 and 10 and desmosomal proteins, disrupting epidermal structure to promote expression of a blistering phenotype. Here, we extend these findings to show that VZV infection upregulates the expression of keratin 15 (KRT15), a marker expressed by basal epidermal keratinocytes and hair follicles stem cells. We demonstrate that KRT15 is essential for VZV replication in the skin, since downregulation of KRT15 inhibits VZV replication in keratinocytes, while KRT15 exogenous overexpression supports viral replication. Importantly, our data show that VZV upregulation of KRT15 depends on the expression of the VZV immediate early gene ORF62. ORF62 is the only regulatory gene that is mutated in the live attenuated VZV vaccine and contains four of the five fixed mutations present in the VZV Oka vaccine. Our data indicate that the mutated vaccine ORF62 is not capable of upregulating KRT15, suggesting that this may contribute to the vaccine attenuation in skin. Taken together our data present a novel association between VZV and KRT15, which may open a new therapeutic window for a topical targeting of VZV replication in the skin via modulation of KRT15.
Collapse
Affiliation(s)
- Cristina Tommasi
- Infection, Immunity and Inflammation Department, University College London GOS Institute of Child Health, London, UK
| | - Ohad Yogev
- Infection and Immunity Department, University College London, London, UK
- Eleven Therapeutics, Cambridge, UK
| | - Michael B Yee
- Department of Ophthalmology and of Molecular Microbiology and Genetics, University of Pittsburgh School of Medicine, Pittsburgh, US
- Krystalbio Inc, Pittsburgh, US
| | - Andriani Drousioti
- Infection, Immunity and Inflammation Department, University College London GOS Institute of Child Health, London, UK
| | - Meleri Jones
- Infection and Immunity Department, University College London, London, UK
- UKHSA, Porton Down, UK
| | - Alice Ring
- Infection and Immunity Department, University College London, London, UK
| | | | - Inga Dry
- Infection and Immunity Department, University College London, London, UK
- The Roslin Institute, Edinburgh, UK
| | - Oscar Atkins
- Infection and Immunity Department, University College London, London, UK
- Francis Crick Institute, London, UK
| | - Aishath S Naeem
- Infection and Immunity Department, University College London, London, UK
- Dana-Farber Cancer Institute, Boston, US
| | - Nisha Kriplani
- Infection Medicine, University of Edinburgh, Edinburgh, UK
| | - Arne N Akbar
- Experimental & Translational Medicine, Division of Medicine, University College London, London, UK
| | - Jürgen G Haas
- Infection Medicine, University of Edinburgh, Edinburgh, UK
| | - Edel A O'Toole
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London, UK
| | - Paul R Kinchington
- Department of Ophthalmology and of Molecular Microbiology and Genetics, University of Pittsburgh School of Medicine, Pittsburgh, US
| | - Judith Breuer
- Infection, Immunity and Inflammation Department, University College London GOS Institute of Child Health, London, UK.
| |
Collapse
|
2
|
Mo ZJ, Huang SJ, Qiu LX, Li CG, Yu XJ, Li MQ, Chen Z, Zhong GH, Pan DQ, Huang LR, Lv BJ, Cui XL, Song QQ, Jia JZ, Han JL, Wang W, Zhu H, Cheng T, Su YY, Li YM, Ye XZ, Wu T, Zhang J, Xia NS. Safety and immunogenicity of a skin- and neuro-attenuated live vaccine for varicella: a randomized, double-blind, controlled, dose-escalation and age de-escalation phase 1 clinical trial. THE LANCET REGIONAL HEALTH - WESTERN PACIFIC 2023. [DOI: 10.1016/j.lanwpc.2023.100707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
|
3
|
Meningitis without Rash after Reactivation of Varicella Vaccine Strain in a 12-Year-Old Immunocompetent Boy. Vaccines (Basel) 2023; 11:vaccines11020309. [PMID: 36851187 PMCID: PMC9964174 DOI: 10.3390/vaccines11020309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/16/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Acute neurologic complications from Varicella-Zoster-Virus reactivation occur in both immunocompromised and immunocompetent patients. In this report, we describe a case of a previously healthy immunocompetent boy who had received two doses of varicella vaccine at 1 and 4 years. At the age of 12 he developed acute aseptic meningitis caused by vaccine-type varicella-zoster-virus without concomitant skin eruptions. VZV-vaccine strain DNA was detected in the cerebrospinal fluid. The patient made a full recovery after receiving intravenous acyclovir therapy. This disease course documents another case of a VZV vaccine-associated meningitis without development of a rash, i.e., a form of VZV infection manifesting as "zoster sine herpete".
Collapse
|
4
|
Jansen MHA, Rondaan C, Legger GE, Minden K, Uziel Y, Toplak N, Maritsi D, van den Berg L, Berbers GAM, Bruijning P, Egert Y, Normand C, Bijl M, Foster HE, Koné-Paut I, Wouters C, Ravelli A, Elkayam O, Wulffraat NM, Heijstek MW. EULAR/PRES recommendations for vaccination of paediatric patients with autoimmune inflammatory rheumatic diseases: update 2021. Ann Rheum Dis 2023; 82:35-47. [PMID: 35725297 DOI: 10.1136/annrheumdis-2022-222574] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/24/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVES Recent insights supporting the safety of live-attenuated vaccines and novel studies on the immunogenicity of vaccinations in the era of biological disease-modifying antirheumatic drugs in paediatric patients with autoimmune/inflammatory rheumatic diseases (pedAIIRD) necessitated updating the EULAR recommendations. METHODS Recommendations were developed using the EULAR standard operating procedures. Two international expert committees were formed to update the vaccination recommendations for both paediatric and adult patients with AIIRD. After a systematic literature review, separate recommendations were formulated for paediatric and adult patients. For pedAIIRD, six overarching principles and seven recommendations were formulated and provided with the level of evidence, strength of recommendation and Task Force level of agreement. RESULTS In general, the National Immunisation Programmes (NIP) should be followed and assessed yearly by the treating specialist. If possible, vaccinations should be administered prior to immunosuppressive drugs, but necessary treatment should never be postponed. Non-live vaccines can be safely given to immunosuppressed pedAIIRD patients. Mainly, seroprotection is preserved in patients receiving vaccinations on immunosuppression, except for high-dose glucocorticoids and B-cell depleting therapies. Live-attenuated vaccines should be avoided in immunosuppressed patients. However, it is safe to administer the measles-mumps-rubella booster and varicella zoster virus vaccine to immunosuppressed patients under specific conditions. In addition to the NIP, the non-live seasonal influenza vaccination should be strongly considered for immunosuppressed pedAIIRD patients. CONCLUSIONS These recommendations are intended for paediatricians, paediatric rheumatologists, national immunisation agencies, general practitioners, patients and national rheumatology societies to attain safe and effective vaccination and optimal infection prevention in immunocompromised pedAIIRD patients.
Collapse
Affiliation(s)
- Marc H A Jansen
- Department of Paediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands .,RITA, European Reference Networks, Brussels, Belgium
| | - Christien Rondaan
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, The Netherlands
| | - Geertje E Legger
- RITA, European Reference Networks, Brussels, Belgium.,Department of Paediatric Rheumatology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
| | - Kirsten Minden
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin, Berlin, Germany.,Epidemiology Unit, German Rheumatism Research Centre, Berlin, Germany
| | - Yosef Uziel
- Paediatric Rheumatology Unit, Department of Paediatrics, Meir Medical Center, Kfar Saba, Israel
| | - Natasa Toplak
- RITA, European Reference Networks, Brussels, Belgium.,Department of Allergology, Rheumatology and Clinical Immunology, University Children's Hospital, Ljubljana, Slovenia
| | - Despoina Maritsi
- Infectious Diseases, Immunology and Rheumatology Unit, Department of Paediatrics, Kyriakou Children's Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Lotte van den Berg
- Dutch JIA Patient and Parent Organisation (JVN), Member of ENCA, Amsterdam, The Netherlands
| | - Guy A M Berbers
- Centre for Infectious Disease Control Netherlands, Laboratory for Infectious Diseases and Screening, National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Patricia Bruijning
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Yona Egert
- European Network Childhood Arthritis (ENCA) Patient Organisation, Judea and Samaria Area, Israel
| | - Christophe Normand
- MCI Secretariat, European Network for Children with Arthritis (ENCA), Geneva, Switzerland
| | - Marc Bijl
- Department of Internal Medicine and Rheumatology, Martini Hospital Groningen, Groningen, The Netherlands
| | - Helen E Foster
- Population and Health Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Isabelle Koné-Paut
- Department of Paediatric Rheumatology and CEREMAIA, Hôpital Bicêtre, AP HP, Université Paris Saclay, Paris, France
| | - Carine Wouters
- Division of Paediatric Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Angelo Ravelli
- RITA, European Reference Networks, Brussels, Belgium.,Department of Rheumatology, Direzione Scientifica, IRCCS Istituto Giannina Gaslini, Genova, Italy.,Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili (DINOGMI), Università degli Studi di Genova, Genoa, Italy
| | - Ori Elkayam
- Department of Rheumatology, Tel Aviv Sourasky Medical Center and the Sackler faculty of medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nicolaas M Wulffraat
- Department of Paediatric Immunology & Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands.,RITA, European Reference Networks, Brussels, Belgium
| | - Marloes W Heijstek
- RITA, European Reference Networks, Brussels, Belgium.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
5
|
Varicella Vaccine: a Molecular Variant That May Contribute to Attenuation. mBio 2022; 13:e0312022. [PMID: 36468883 PMCID: PMC9765671 DOI: 10.1128/mbio.03120-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2022] Open
Abstract
Varicella was troublesome when varicella vaccine (vOka) was licensed in the United States. Varicella's yearly death toll was ~100, indirect costs were massive, and varicella threatened immunocompromised children. Since licensure, varicella has almost disappeared; nevertheless, vOka attenuation has lacked a molecular explanation. Sadaoka et al. (T. Sadaoka, D. P. Depledge, L. Rajbhandari, J. Breuer, et al., mBio 13:e0186422, 2022, https://doi.org/10.1128/mbio.01864-22), however, have now identified 6 core single nucleotide polymorphisms (SNPs), which singly or in combination may contribute to VOka attenuation; moreover, they found a predominant variant allele of vOka encoding the viral glycoprotein gB that results in glutamine instead of arginine at amino acid 699. This change impairs fusion activity and the ability of varicella-zoster virus (VZV) to infect human neurons from axon terminals. Molecular virological studies of vOka are reassuring in suggesting that reversion to virulence is unlikely and should also help assuage current fears about VZV vaccination and alleviate unanticipated future problems. The impressive work of Sadaoka et al. thus represents an auspicious advance in knowledge.
Collapse
|
6
|
Shapiro ED, Marin M. The Effectiveness of Varicella Vaccine: 25 Years of Postlicensure Experience in the United States. J Infect Dis 2022; 226:S425-S430. [PMID: 36265844 DOI: 10.1093/infdis/jiac299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
We summarize studies of varicella vaccine's effectiveness for prevention of varicella and lessons learned during the first 25 years of the varicella vaccination program in the United States. One dose of varicella vaccine provided moderate protection (82%-85%) against varicella of any severity and high protection (100%) against severe varicella, with some waning of protection over time. The 1-dose program (1995-2006) had a substantial impact on the incidence both of varicella and of severe outcomes (71%-90% decrease) although it did not prevent low-level community transmission and some outbreaks continued to occur in highly vaccinated populations. Two doses of varicella vaccine improved the vaccine's effectiveness by at least 10% against varicella of any severity, with further declines in the incidence both of varicella and of severe outcomes as well as in both number and size of outbreaks. There is no evidence for waning of the effectiveness of 2 doses of the vaccine.
Collapse
Affiliation(s)
- Eugene D Shapiro
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Mona Marin
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
7
|
Arvin AM. Insights From Studies of the Genetics, Pathogenesis, and Immunogenicity of the Varicella Vaccine. J Infect Dis 2022; 226:S385-S391. [PMID: 36265853 DOI: 10.1093/infdis/jiac278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
While the varicella vaccine was created with approaches established for other live attenuated viral vaccines, novel methods to probe virus-host interactions have been used to explore the genetics, pathogenesis, and immunogenicity of the vaccine compared to wild-type varicella-zoster virus (VZV). As summarized here, a mechanism-based understanding of the safety and efficacy of the varicella vaccine has been achieved through these investigations.
Collapse
Affiliation(s)
- Ann M Arvin
- Departments of Pediatrics and Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
8
|
Pawaskar M, Siddiqui MK, Takyar J, Sharma A, Fergie J. Relative efficacy of varicella vaccines: network meta-analysis of randomized controlled trials. Curr Med Res Opin 2022; 38:1772-1782. [PMID: 35713564 DOI: 10.1080/03007995.2022.2091334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Although varicella vaccination is highly effective, no head-to-head randomized controlled trials (RCTs) have compared the efficacy of different vaccine formulations. This study assessed the relative efficacy of different varicella vaccines using network meta-analysis (NMA). METHODS We estimated the relative efficacies of varicella vaccines and dosing regimens from RCTs using Bayesian NMA. Modeling-based time-series NMA (MBNMA) was performed, accounting for differences in time since vaccination, to extrapolate long-term vaccine efficacy (VE). RESULTS Eight RCTs were included based on systematic review of biomedical databases. Efficacy data were reported for four varicella-containing vaccines: Varivax (V-MSD, one and two dose), Varilrix (V-GSK, one dose), Priorix-Tetra (MMRV-GSK, one dose), and Sinovac (V-Sinovac, one dose). All varicella vaccines were effective versus no vaccination. Two-dose V-MSD (98.29%, 95% credible interval [CrI] 96.08-99.23) showed significantly higher VE versus all one-dose varicella-containing vaccines, but no significant difference versus two-dose MMRV-GSK (95.19%, 95% CrI 90.3-97.63). Two-dose MMRV-GSK showed higher VE than one-dose V-GSK (66.47%; 95% CrI 43.02-79.43), but no significant differences in VE versus one-dose V-MSD or one-dose V-Sinovac. In one-dose comparisons, V-MSD showed significantly higher VE (93.09%, 95% CrI 89.13-95.96) than V-GSK, but no significant difference versus V-Sinovac (89.22%; 95% CrI 67.1-96.5). MBNMA indicated that protection against varicella was sustained without waning over the 10 year follow-up. CONCLUSIONS Our study reported higher VE for two-dose V-MSD and MMRV-GSK. Among one-dose formulations, one-dose V-MSD was more efficacious than one-dose V-GSK. Policymakers should take into consideration differences in VE when implementing one- versus two-dose strategies in universal vaccination programs.
Collapse
Affiliation(s)
- Manjiri Pawaskar
- Center for Observational and Real-World Evidence, Merck & Co., Inc., Rahway, NJ, USA
| | | | - Jitender Takyar
- Parexel Regulatory & Access, Parexel International, Mohali, India
| | - Akanksha Sharma
- Center for Observational and Real-World Evidence, Merck & Co., Inc., Rahway, NJ, USA
| | - Jaime Fergie
- Department of Pediatrics, Driscoll Children's Hospital, Corpus Christi, TX, USA
| |
Collapse
|
9
|
Candido KL, Eich CR, de Fariña LO, Kadowaki MK, da Conceição Silva JL, Maller A, Simão RDCG. Spike protein of SARS-CoV-2 variants: a brief review and practical implications. Braz J Microbiol 2022; 53:1133-1157. [PMID: 35397075 PMCID: PMC8994061 DOI: 10.1007/s42770-022-00743-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/21/2022] [Indexed: 12/24/2022] Open
Abstract
The scientific community has been alarmed by the possible immunological evasion, higher infectivity, and severity of disease caused by the newest variants of SARS-CoV-2. The spike protein has an important role in the cellular invasion of viruses and is the target of several vaccines and therapeutic resources, such as monoclonal antibodies. In addition, some of the most relevant mutations in the different variants are on the spike (S) protein gene sequence that leads to structural alterations in the predicted protein, thus causing concern about the protection mediated by vaccines against these new strains. The present review highlights the most recent knowledge about COVID-19 and vaccines, emphasizing the different spike protein structures of SARS-CoV-2 and updating the reader about the emerging viral variants and their classifications, the more common viral mutations described and their distribution in Brazil. It also compiles a table with the most recent knowledge about all of the Omicron spike mutations.
Collapse
Affiliation(s)
- Kattlyn Laryssa Candido
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| | - Caio Ricardo Eich
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| | - Luciana Oliveira de Fariña
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| | - Marina Kimiko Kadowaki
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| | - José Luis da Conceição Silva
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| | - Alexandre Maller
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| | - Rita de Cássia Garcia Simão
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| |
Collapse
|
10
|
Levin MJ, Weinberg A. Immune Responses to Varicella-Zoster Virus Vaccines. Curr Top Microbiol Immunol 2022; 438:223-246. [PMID: 35102438 DOI: 10.1007/82_2021_245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The live attenuated varicella vaccine is intended to mimic the tempo and nature of the humoral and cell-mediated immune responses to varicella infection. To date, two doses of varicella vaccine administered in childhood have been very effective in generating varicella-zoster virus (VZV) immune responses that prevent natural infection for at least several decades. After primary infection, the infecting VZV establishes latency in sensory and cranial nerve ganglia with the potential to reactivate and cause herpes zoster. Although, the immune responses developed during varicella are important for preventing herpes zoster they wane with increasing age (immune senescence) or with the advent of immune suppression. Protection can be restored by increasing cell-mediated immune responses with two doses of an adjuvanted recombinant VZV glycoprotein E vaccine that stimulates both VZV-and gE-specific immunity. This vaccine provides ~85-90% protection against herpes zoster for 7-8 years (to date).
Collapse
Affiliation(s)
- Myron J Levin
- Departments of Pediatrics and Medicine, University of Colorado Denver School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Adriana Weinberg
- Departments of Pediatrics, Medicine, and Pathology, University of Colorado Denver School of Medicine, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
11
|
Safety surveillance of varicella vaccine using tree-temporal scan analysis. Vaccine 2021; 39:6378-6384. [PMID: 34561139 DOI: 10.1016/j.vaccine.2021.09.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/03/2021] [Accepted: 09/13/2021] [Indexed: 11/20/2022]
Abstract
IMPORTANCE Passive surveillance systems are susceptible to the under-reporting of adverse events (AE) and a lack of information pertaining to vaccinated populations. Conventional active surveillance focuses on predefined AEs. Advanced data mining tools could be used to identify unusual clusters of potential AEs after vaccination. OBJECTIVE To assess the feasibility of a novel tree-based statistical approach to the identification of AE clustering following the implementation of a varicella vaccination program among one-year-olds. SETTING AND PARTICIPANTS This nationwide safety surveillance was based on data from the Taiwan National Health Insurance database and National Immunization Information System for the period 2004 through 2014. The study population was children aged 12-35 months who received the varicella vaccine. EXPOSURE First-dose varicella vaccine. OUTCOMES AND MEASURES All incident ICD-9-CM diagnoses (emergency or inpatient departments) occurring 1-56 days after the varicella vaccination were classified within a hierarchical system of diagnosis categories using Multi-Level Clinical Classifications Software. A self-controlled tree-temporal data mining tool was then used to explore the incidence of AE clustering with a variety of potential risk intervals. The comparison interval consisted of days in the 56-day follow-up period that fell outside the risk interval. RESULTS Among 1,194,189 varicella vaccinees with no other same-day vaccinations, nine diagnoses with clustering features were categorized into four safety signals: fever on days 1-6 (attributable risk [AR] 38.5 per 100,000, p < 0.001), gastritis and duodenitis on days 1-2 (AR 5.9 per 100,000, p < 0.001), acute upper respiratory infection on days 1-5 (AR 11.0 per 100,000, p = 0.006), and varicella infection on days 1-9 (AR 2.7 per 100,000, p < 0.001). These safety profiles and their corresponding risk intervals have been identified in previous safety surveillance studies. CONCLUSIONS Unexpected clusters of AEs were not detected after the mass administration of childhood varicella vaccines in Taiwan. The tree-temporal statistical method is a feasible approach to the safety surveillance of vaccines in populations of young children.
Collapse
|
12
|
Abstract
Michiaki Takahashi developed the live attenuated varicella vaccine in 1974 . This was the first, and is still the only, herpesvirus vaccine. Early studies showed promise, but the vaccine was rigorously tested on immunosuppressed patients because of their high risk of fatal varicella; vaccination proved to be lifesaving. Subsequently, the vaccine was found to be safe and effective in healthy children. Eventually, varicella vaccine became a component of measles mumps rubella vaccine, 2 doses of which are administered in the USA to ~90% of children. The incidence of varicella has dropped dramatically in the USA since vaccine-licensure in 1995. Varicella vaccine is also associated with a decreased incidence of zoster and is protective for susceptible adults. Today, immunocompromised individuals are protected against varicella due to vaccine-induced herd immunity. Latent infection with varicella zoster virus occurs after vaccination; however, the vaccine strain is impaired for its ability to reactivate.
Collapse
Affiliation(s)
- Anne A Gershon
- Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Michael D Gershon
- Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | | |
Collapse
|
13
|
Akpo EIH, Cristeau O, Hunjan M, Casabona G. Reply to Pawaskar et al. Clin Infect Dis 2021; 73:936-937. [PMID: 34492691 PMCID: PMC8423474 DOI: 10.1093/cid/ciab046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
14
|
Kuter BJ, Offit PA, Poland GA. The development of COVID-19 vaccines in the United States: Why and how so fast? Vaccine 2021; 39:2491-2495. [PMID: 33824043 PMCID: PMC7997594 DOI: 10.1016/j.vaccine.2021.03.077] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 01/19/2023]
Affiliation(s)
- Barbara J Kuter
- Vaccine Education Center, Children's Hospital of Philadelphia, United States
| | - Paul A Offit
- Vaccine Education Center, Children's Hospital of Philadelphia, United States
| | | |
Collapse
|
15
|
Immunogenicity and safety profiles of a new MAV/06 strain varicella vaccine in healthy children: A multinational, multicenter, randomized, double-blinded, active-controlled phase III study. Vaccine 2021; 39:1758-1764. [PMID: 33627245 DOI: 10.1016/j.vaccine.2021.02.013] [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: 08/23/2020] [Revised: 01/12/2021] [Accepted: 02/06/2021] [Indexed: 11/22/2022]
Abstract
Immunization is the most effective preventive strategy against varicella. While the Oka strain is commonly used for varicella vaccination worldwide, Korea widely uses the MAV/06 strain. A new live attenuated MAV/06 strain varicella vaccine (MG1111), which uses the new cell line Medical Research Council-5 for better viral propagation, was developed. MG1111 was approved by Korean health authorities. Here, we report the results of phase III, randomized, double-blind, multicenter study conducted in Korea and Thailand, which compared the immunogenicity and safety profiles of MG1111 versus the control vaccine, VarivaxTM. In total, 515 healthy children (12 month-12 years) were randomized 1:1 to receive either the MG1111 or control vaccine (MG1111: 258, Control: 257). The seroconversion rate (SCR) and geometric mean titer (GMT) were measured using the fluorescent antibody to membrane antigen (FAMA) test. The MG1111 group achieved a SCR of 97.9% (95% CI: 95.2-99.3) after vaccination. The lower limit of 95% CI for SCR difference (MG1111-VarivaxTM) was -4.0%, which was higher than the specified non-inferiority margin of -10%. Further, the GMT of the MG1111 increased from 2.0 to 74.2 (95% CI: 65.0-84.8) and the lower limits of the 95% CI for post-vaccination GMT ratios (MG1111/VarivaxTM) were 0.55 higher than the specified parameter of 0.5. Therefore, the MG1111 group was not statistically inferior to the control vaccine group in terms of SCR and GMT. Furthermore, the MG1111 and control vaccine groups were not significantly different in the percentage of participants showing adverse events-solicited, local, or systemic during 43-day period of observation and serious adverse events during 6 month of observation. The present results indicate that MG1111was not immunologically inferior to VarivaxTM, and safety profiles of MG1111 are similar to those of VarivaxTM.
Collapse
|
16
|
Bernstein DI, Cardin RD, Smith GA, Pickard GE, Sollars PJ, Dixon DA, Pasula R, Bravo FJ. The R2 non-neuroinvasive HSV-1 vaccine affords protection from genital HSV-2 infections in a guinea pig model. NPJ Vaccines 2020; 5:104. [PMID: 33298966 PMCID: PMC7648054 DOI: 10.1038/s41541-020-00254-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 10/06/2020] [Indexed: 02/01/2023] Open
Abstract
Herpes simplex virus (HSV) infections are common and can cause severe illness but no vaccine is currently available. The recent failure of subunit HSV vaccines has highlighted the need for vaccines that present a diverse array of antigens, including the development of next-generation live-attenuated vaccines. However, most attenuated HSV strains propagate poorly, limiting their ability to elicit protective immune responses. A live-attenuated vaccine that replicates in non-neural tissue but is ablated for transmission into the nervous system may elicit protective immune responses without evoking neurologic complications or establishing life-long infections. Initial studies of R2, a live-attenuated vaccine that is engineered to be unable to invade the nervous system, used the guinea pig genital HSV model to evaluate the ability of R2 to replicate at the site of inoculation, cause disease and infect neural tissues. R2 was then evaluated as a vaccine using three routes of inoculation: intramuscular (IM), intradermal (ID) and intravaginal (IVag) and compared to IM administered gD2+MPL/Alum vaccine in the same model. R2 replicated in the genital tract but did not produce acute or recurrent disease and did not infect the neural tissue. The R2 vaccine-induced neutralizing antibody and decreased the severity of acute and recurrent HSV-2 disease as well as recurrent shedding. The ID route was the most effective. ID administered R2 was more effective than gD2+MPL/Alum at inducing neutralizing antibody, suppressing acute disease, and acute vaginal virus replication. R2 was especially more effective at reducing recurrent virus shedding, the most common source of HSV transmission. The live-attenuated prophylactic HSV vaccine, R2, was effective in the guinea pig model of genital HSV-2 especially when administered by the ID route. The use of live-attenuated HSV vaccines that robustly replicate in mucosal tissues but are ablated for neuroinvasion offers a promising approach for HSV vaccines.
Collapse
Affiliation(s)
- David I Bernstein
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA.
| | - Rhonda D Cardin
- School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Gregory A Smith
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Gary E Pickard
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska, Lincoln, NE, USA
| | - Patricia J Sollars
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska, Lincoln, NE, USA
| | - David A Dixon
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Rajamouli Pasula
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Fernando J Bravo
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
17
|
Castro MC, Rojas P. Preventive effectiveness of varicella vaccine in healthy unexposed patients. Medwave 2020; 20:e7983. [PMID: 32759893 DOI: 10.5867/medwave.2020.06.7982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 11/28/2019] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Chickenpox is an infectious disease caused by varicella-zoster virus. Varicella vaccine is conventionally used for its prevention, and its administration seeks to reduce the onset of the disease and complications associated. However, there is still controversy about its effectiveness. METHODS We searched in Epistemonikos, the largest database of systematic reviews in health, which is maintained by screening multiple information sources, including MEDLINE, EMBASE, Cochrane, among others. We extracted data from the systematic reviews, reanalyzed data of primary studies, conducted a meta-analysis and generated a summary of findings table using the GRADE approach. RESULTS AND CONCLUSIONS We identified two systematic reviews including 16 studies overall, of which three were randomized trials. We concluded that the varicella vaccine decreases the risk of contracting the disease in the long term and probably reduces the risk of developing the disease in the short term in healthy unexposed patients. Nevertheless, the vaccination increases the occurrence of local reactions 48 hours after its administration and probably increases the presence of fever and chickenpox-like rash.
Collapse
Affiliation(s)
- María Catalina Castro
- Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Proyecto Epistemonikos, Santiago, Chile
| | - Pamela Rojas
- Proyecto Epistemonikos, Santiago, Chile; Departamento de Medicina Familiar del Niño, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile. . Address: Centro Evidencia UC, Pontificia Universidad Católica de Chile, Diagonal Paraguay 476, Santiago, Chile
| |
Collapse
|
18
|
Liu W, Cui Y, Wang C, Li Z, Gong D, Dai X, Bi GQ, Sun R, Zhou ZH. Structures of capsid and capsid-associated tegument complex inside the Epstein-Barr virus. Nat Microbiol 2020; 5:1285-1298. [PMID: 32719506 DOI: 10.1038/s41564-020-0758-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 06/19/2020] [Indexed: 12/20/2022]
Abstract
As the first discovered human cancer virus, Epstein-Barr virus (EBV) causes Burkitt's lymphoma and nasopharyngeal carcinoma. Isolating virions for determining high-resolution structures has been hindered by latency-a hallmark of EBV infection-and atomic structures are thus available only for recombinantly expressed EBV proteins. In the present study, by symmetry relaxation and subparticle reconstruction, we have determined near-atomic-resolution structures of the EBV capsid with an asymmetrically attached DNA-translocating portal and capsid-associated tegument complexes from cryogenic electron microscopy images of just 2,048 EBV virions obtained by chemical induction. The resulting atomic models reveal structural plasticity among the 20 conformers of the major capsid protein, 2 conformers of the small capsid protein (SCP), 4 conformers of the triplex monomer proteins and 2 conformers of the triplex dimer proteins. Plasticity reaches the greatest level at the capsid-tegument interfaces involving SCP and capsid-associated tegument complexes (CATC): SCPs crown pentons/hexons and mediate tegument protein binding, and CATCs bind and rotate all five periportal triplexes, but notably only about one peri-penton triplex. These results offer insights into the EBV capsid assembly and a mechanism for recruiting cell-regulating factors into the tegument compartment as 'cargoes', and should inform future anti-EBV strategies.
Collapse
Affiliation(s)
- Wei Liu
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.,Center for Integrative Imaging, Hefei National Laboratory for Physical Sciences at the Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yanxiang Cui
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Caiyan Wang
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.,International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zihang Li
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Danyang Gong
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Therapeutics Discovery, Amgen Research, Amgen Inc., Thousand Oaks, CA, USA
| | - Xinghong Dai
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Guo-Qiang Bi
- Center for Integrative Imaging, Hefei National Laboratory for Physical Sciences at the Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Ren Sun
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Z Hong Zhou
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA. .,Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Zeng WB, Zhang F, Cheng S, Sun JY, Shen H, Luo MH. Concerns on Vaccine against Varicella Caused by Varicella-Zoster Virus Infection. Virol Sin 2020; 36:159-162. [PMID: 32468419 DOI: 10.1007/s12250-020-00231-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/17/2020] [Indexed: 12/30/2022] Open
Affiliation(s)
- Wen-Bo Zeng
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Fukun Zhang
- Changchun Keygen Biological Products Co. Ltd, Changchun, 130000, China
| | - Shuang Cheng
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jin-Yan Sun
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Hongjie Shen
- Changchun Keygen Biological Products Co. Ltd, Changchun, 130000, China.
| | - Min-Hua Luo
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
20
|
Liu CH, Yeh YC, Huang WT, Chie WC, Chan KA. Assessment of pre-specified adverse events following varicella vaccine: A population-based self-controlled risk interval study. Vaccine 2020; 38:2495-2502. [PMID: 32046891 DOI: 10.1016/j.vaccine.2020.01.090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 12/20/2019] [Accepted: 01/30/2020] [Indexed: 10/25/2022]
Abstract
BACKGROUND Clinical trials and spontaneous reporting systems have revealed rare but biologically plausible adverse events following varicella immunization. Few post-marketing controlled studies have been conducted to assess the relationship between the varicella vaccine and these outcomes. OBJECTIVES To evaluate the risk of pneumonia, idiopathic thrombocytopenic purpura (ITP), meningitis, encephalitis and ischemic stroke following varicella immunization. MATERIALS AND METHODS This nationwide observational study was based on Taiwan National Health Insurance data and National Immunization Information System from 2004 through 2014. Primary analysis included children aged 12-35 months who received the single varicella vaccine on the date of administration. The self-controlled risk interval design compared the incidence of pre-specified outcomes during a risk interval of 1-42 days post-vaccination and a control interval of 43-84 days. The outcomes of interest were defined as admitted pneumonia, ITP, meningitis, encephalitis, and ischemic stroke, as well as fracture as a negative control. Conditional Poisson regression was used to assess the incidence rate ratio (aIRR) with adjustments for age and seasonal effects. RESULTS Among 1,194,189 children, who receiving the varicella vaccine, there was no observed increase in the risk for ITP (aIRR 1.00; 95% CI, 0.76-1.33), meningitis (aIRR 1.21; 95% CI, 0.49-2.95), encephalitis (aIRR 1.00; 95% CI, 0.62-1.60), or ischemic stroke (aIRR 1.24; 95% CI, 0.31-4.95). A clustering feature with pneumonia occurred during days 36-42 post-vaccination (aIRR 1.10; 95% CI, 1.02-1.18). An increase in the risk for ITP was observed in children receiving the varicella and MMR vaccines concomitantly (aIRR 1.70; 95% CI, 1.19-2.43), but not among those receiving the varicella vaccine only. CONCLUSIONS We detected a small risk of incidental pneumonia associated with varicella vaccine in the 6th week after immunization. There was no increase in the risk of other pre-specified adverse events.
Collapse
Affiliation(s)
- Chia-Hung Liu
- Graduate Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Family Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chun Yeh
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Wan-Ting Huang
- Taiwan Centers for Disease Control, Ministry of Health and Welfare, Taipei, Taiwan.
| | - Wei-Chu Chie
- Graduate Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - K Arnold Chan
- Health Data Research Center, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
21
|
Affiliation(s)
- Stewart Sell
- Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY, USA
| |
Collapse
|
22
|
Wilmschen S, Schmitz JE, Kimpel J. Viral Vectors for the Induction of Broadly Neutralizing Antibodies against HIV. Vaccines (Basel) 2019; 7:vaccines7030119. [PMID: 31546894 PMCID: PMC6789710 DOI: 10.3390/vaccines7030119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/12/2019] [Accepted: 09/16/2019] [Indexed: 01/10/2023] Open
Abstract
Extensive research on generating an efficient HIV vaccine is ongoing. A major aim of HIV vaccines is the induction of long-lasting, broadly neutralizing antibodies (bnAbs) that can confer sterile immunity for a prolonged period of time. Several strategies have been explored to reach this goal, i.e. protein immunization, DNA, or viral vectors, or a combination thereof. In this review, we give an overview of approaches using viral vectors for the induction of HIV-specific bnAbs. Many pre-clinical studies were performed using various replication-competent and -incompetent vectors. Amongst them, poxviral and adenoviral vectors were the most prevalent ones. In many studies, viral vectors were combined with a DNA prime or a protein boost. However, neutralizing antibodies were mainly induced against the homologous HIV-1 vaccine strain or tier 1 viruses, and in rare cases, against tier 2 viruses, indicating the need for improved antigens and vaccination strategies. Furthermore, we also review next generation Env antigens that are currently being used in protein vaccination approaches and point out how they could be utilized in viral vectors.
Collapse
Affiliation(s)
- Sarah Wilmschen
- Division of Virology, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Joern E Schmitz
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Janine Kimpel
- Division of Virology, Medical University of Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
23
|
Marin M, Leung J, Gershon AA. Transmission of Vaccine-Strain Varicella-Zoster Virus: A Systematic Review. Pediatrics 2019; 144:peds.2019-1305. [PMID: 31471448 PMCID: PMC6957073 DOI: 10.1542/peds.2019-1305] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/03/2019] [Indexed: 11/24/2022] Open
Abstract
CONTEXT Live vaccines usually provide robust immunity but can transmit the vaccine virus. OBJECTIVE To assess the characteristics of secondary transmission of the vaccine-strain varicella-zoster virus (Oka strain; vOka) on the basis of the published experience with use of live varicella and zoster vaccines. DATA SOURCES Systematic review of Medline, Embase, the Cochrane Library, Cumulative Index to Nursing and Allied Health Literature, and Scopus databases for articles published through 2018. STUDY SELECTION Articles that reported original data on vOka transmission from persons who received vaccines containing the live attenuated varicella-zoster virus. DATA EXTRACTION We abstracted data to describe vOka transmission by index patient's immune status, type (varicella or herpes zoster) and severity of illness, and whether transmission was laboratory confirmed. RESULTS Twenty articles were included. We identified 13 patients with vOka varicella after transmission from 11 immunocompetent varicella vaccine recipients. In all instances, the vaccine recipient had a rash: 6 varicella-like and 5 herpes zoster. Transmission occurred mostly to household contacts. One additional case was not considered direct transmission from a vaccine recipient, but the mechanism was uncertain. Transmission from vaccinated immunocompromised children also occurred only if the vaccine recipient developed a rash postvaccination. Secondary cases of varicella caused by vOka were mild. LIMITATIONS It is likely that other vOka transmission cases remain unpublished. CONCLUSIONS Healthy, vaccinated persons have minimal risk for transmitting vOka to contacts and only if a rash is present. Our findings support the existing recommendations for routine varicella vaccination and the guidance that persons with vaccine-related rash avoid contact with susceptible persons at high risk for severe varicella complications.
Collapse
Affiliation(s)
- Mona Marin
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia; and
| | - Jessica Leung
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Anne A. Gershon
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| |
Collapse
|
24
|
Interferon Gamma Inhibits Varicella-Zoster Virus Replication in a Cell Line-Dependent Manner. J Virol 2019; 93:JVI.00257-19. [PMID: 30918075 DOI: 10.1128/jvi.00257-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/21/2019] [Indexed: 01/29/2023] Open
Abstract
The major immediate early 62 (IE62) protein of varicella-zoster virus (VZV) is delivered to newly infected cell nuclei, where it initiates VZV replication by transactivating viral immediate early (IE), early (E), and late (L) genes. Interferon gamma (IFN-γ) is a potent cytokine produced following primary VZV infection. Furthermore, VZV reactivation correlates with a decline in IFN-γ-producing immune cells. Our results showed that treatment with 20 ng/ml of IFN-γ completely reduced intracellular VZV yield in A549 lung epithelial cells, MRC-5 lung fibroblasts, and ARPE-19 retinal epithelial cells at 4 days post-VZV infection. However, IFN-γ reduced virus yield only 2-fold in MeWo melanoma cells compared to that of untreated cells. IFN-β significantly inhibited VZV replication in both ARPE-19 and MeWo cells. In luciferase assays with VZV open reading frame 61 (ORF61) promoter reporter plasmid, IFN-γ abrogated the transactivation activity of IE62 by 95%, 97%, and 89% in A549, ARPE-19, and MRC-5 cells, respectively. However, IFN-γ abrogated IE62's transactivation activity by 16% in MeWo cells, indicating that IFN-γ inhibits VZV replication as well as IE62-mediated transactivation in a cell line-dependent manner. The expression of VZV IE62 and ORF63 suppressed by IFN-γ was restored by JAK1 inhibitor treatment, indicating that the inhibition of VZV replication is mediated by JAK/STAT1 signaling. In the presence of IFN-γ, knockdown of interferon response factor 1 (IRF1) increased VZV replication. Ectopic expression of IRF1 reduced VZV yields 4,000-fold in MRC-5 and ARPE-19 cells but 3-fold in MeWo cells. These results suggest that IFN-γ blocks VZV replication by inhibiting IE62 function in a cell line-dependent manner.IMPORTANCE Our results showed that IFN-γ significantly inhibited VZV replication in a cell line-dependent manner. IFN-γ inhibited VZV gene expression after the immediate early stage of infection and abrogated IE62-mediated transactivation. These results suggest that IFN-γ blocks VZV replication by inhibiting IE62 function in a cell line-dependent manner. Understanding the mechanisms by which IFN-γ plays a role in VZV gene programming may be important in determining the tissue restriction of VZV.
Collapse
|
25
|
Sullivan NL, Eberhardt CS, Wieland A, Vora KA, Pulendran B, Ahmed R. Understanding the immunology of the Zostavax shingles vaccine. Curr Opin Immunol 2019; 59:25-30. [PMID: 30970291 DOI: 10.1016/j.coi.2019.02.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 11/17/2022]
Abstract
Zostavax is a live-attenuated varicella zoster virus (VZV) vaccine recommended for use in adults >50 years of age to prevent shingles. The main risk factor for the development of shingles is age, which correlates with decreasing cell-mediated immunity. These data suggest a predominant role of T cell immunity in controlling VZV latency. However, other components of the immune system may also contribute. In this review, we will discuss how the immune system responds to Zostavax, focusing on recent studies examining innate immunity, transcriptomics, metabolomics, cellular, and humoral immunity.
Collapse
Affiliation(s)
- Nicole L Sullivan
- MRL, Department of Infectious Diseases and Vaccines, Merck & Co., Inc., Kenilworth, New Jersey, USA.
| | - Christiane S Eberhardt
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA; Center for Vaccinology and Neonatal Immunology, Department of Pediatrics and Pathology-Immunology, University Hospitals of Geneva and Faculty of Medicine, University of Geneva, Switzerland
| | - Andreas Wieland
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Kalpit A Vora
- MRL, Department of Infectious Diseases and Vaccines, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Department of Pathology, Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
26
|
Ong CEB, Lyons AB, Woods GM, Flies AS. Inducible IFN-γ Expression for MHC-I Upregulation in Devil Facial Tumor Cells. Front Immunol 2019; 9:3117. [PMID: 30692995 PMCID: PMC6340284 DOI: 10.3389/fimmu.2018.03117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/17/2018] [Indexed: 12/31/2022] Open
Abstract
The Tasmanian devil facial tumor (DFT) disease has led to an 80% reduction in the wild Tasmanian devil (Sarcophilus harrisii) population since 1996. The limited genetic diversity of wild devils and the lack of MHC-I expression on DFT cells have been implicated in the lack of immunity against the original DFT clonal cell line (DFT1). Recently, a second transmissible tumor of independent origin (DFT2) was discovered. Surprisingly, DFT2 cells do express MHC-I, but DFT2 cells appear to be on a trajectory for reduced MHC-I expression in vivo. Thus, much of the ongoing vaccine-development efforts and conservation plans have focused on MHC-I. A major limitation in conservation efforts is the lack of species-specific tools to understand Tasmanian devil gene function and immunology. To help fill this gap, we developed an all-in-one Tet-Off vector system to regulate expression of IFN-γ in DFT cells (DFT1.Tet/IFN-γ). IFN-γ can have negative effects on cell proliferation and viability; thus, doxycycline was used to suppress IFN-γ production whilst DFT1.Tet/IFN-γ cells were expanded in cell culture. Induction of IFN-γ following removal of doxycycline led to upregulation of MHC-I but also the inhibitory checkpoint molecule PD-L1. Additionally, DFT1.Tet/IFN-γ cells were capable of stimulating MHC-I upregulation on bystander wild type DFT cells in co-culture assays in vitro. This system represents a major step forward in DFT disease immunotherapy and vaccine development efforts, and ability to understand gene function in devils. Importantly, the techniques are readily transferable for testing gene function in DFT2 cells and other non-traditional species.
Collapse
Affiliation(s)
- Chrissie E B Ong
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Alan Bruce Lyons
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Gregory M Woods
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Andrew S Flies
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
27
|
Bernstein DI, Pullum DA, Cardin RD, Bravo FJ, Dixon DA, Kousoulas KG. The HSV-1 live attenuated VC2 vaccine provides protection against HSV-2 genital infection in the guinea pig model of genital herpes. Vaccine 2019; 37:61-68. [DOI: 10.1016/j.vaccine.2018.11.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/09/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022]
|
28
|
Quinn HE, Gidding HF, Marshall HS, Booy R, Elliott EJ, Richmond P, Crawford N, McIntyre PB, Macartney KK. Varicella vaccine effectiveness over 10 years in Australia; moderate protection from 1-dose program. J Infect 2018; 78:220-225. [PMID: 30528868 DOI: 10.1016/j.jinf.2018.11.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 09/25/2018] [Accepted: 11/18/2018] [Indexed: 10/27/2022]
Abstract
OBJECTIVES To examine the impact of Australia's single dose infant varicella vaccination program, we assessed single dose varicella vaccine effectiveness (VE) in preventing hospitalised disease using two methods. METHODS Clinically confirmed varicella cases from the Paediatric Active Enhanced Disease Surveillance (PAEDS) sentinel network were age-matched to 20 controls obtained from the Australian Immunisation Register. Conditional logistic regression models were used to estimate VE and compared with estimates obtained using our second approach. RESULTS There were 78 hospitalised varicella cases during the post vaccine introduction period from January 2008 to December 2015, who were eligible for funded varicella vaccination. Median age at onset was 4.5 years and more than half (59%) were vaccinated. The majority of children received one vaccine brand (Varilrix, GSK). The estimated case-control VE for one dose of vaccine against hospitalised varicella was 64.7% (95% CI: 43.3-78.0%); estimates using the screening method were not significantly different. Exclusion of children who were immunocompromised did not significantly alter VE estimates. CONCLUSIONS Although Australia's program has impacted on the burden of varicella disease, single dose VE against varicella hospitalisation is only moderate. Greater reductions in varicella disease could potentially be achieved by incorporation of a second vaccine dose into the program to minimise breakthrough disease and interrupt virus circulation.
Collapse
Affiliation(s)
- Helen E Quinn
- National Centre for Immunisation Research and Surveillance of Vaccine Preventable Diseases (NCIRS), Children's Hospital at Westmead, Cnr Hawkesbury Road and Hainsworth Street, Westmead, NSW 2145, Australia; Discipline of Child and Adolescent Health, University of Sydney, Sydney, Australia.
| | - Heather F Gidding
- National Centre for Immunisation Research and Surveillance of Vaccine Preventable Diseases (NCIRS), Children's Hospital at Westmead, Cnr Hawkesbury Road and Hainsworth Street, Westmead, NSW 2145, Australia; School of Public Health and Community Medicine, UNSW Medicine, University of New South Wales, Sydney, Australia
| | - Helen S Marshall
- Vaccinology and Immunology Research Trials Unit, Women's and Children's Health Network, Adelaide, Australia; Robinson Research Institute and School of Medicine, University of Adelaide, Adelaide, Australia
| | - Robert Booy
- National Centre for Immunisation Research and Surveillance of Vaccine Preventable Diseases (NCIRS), Children's Hospital at Westmead, Cnr Hawkesbury Road and Hainsworth Street, Westmead, NSW 2145, Australia; Discipline of Child and Adolescent Health, University of Sydney, Sydney, Australia; Children's Hospital at Westmead, Sydney, Australia
| | - Elizabeth J Elliott
- Discipline of Child and Adolescent Health, University of Sydney, Sydney, Australia; Children's Hospital at Westmead, Sydney, Australia; Australian Paediatric Surveillance Unit, Westmead, Australia
| | - Peter Richmond
- Wesfarmer's Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Australia
| | - Nigel Crawford
- Murdoch Children's Research Institute, Parkville, Australia; Royal Children's Hospital, Melbourne, Australia; Department of Paediatrics, The University of Melbourne, Australia
| | - Peter B McIntyre
- National Centre for Immunisation Research and Surveillance of Vaccine Preventable Diseases (NCIRS), Children's Hospital at Westmead, Cnr Hawkesbury Road and Hainsworth Street, Westmead, NSW 2145, Australia; Discipline of Child and Adolescent Health, University of Sydney, Sydney, Australia; Children's Hospital at Westmead, Sydney, Australia
| | - Kristine K Macartney
- National Centre for Immunisation Research and Surveillance of Vaccine Preventable Diseases (NCIRS), Children's Hospital at Westmead, Cnr Hawkesbury Road and Hainsworth Street, Westmead, NSW 2145, Australia; Discipline of Child and Adolescent Health, University of Sydney, Sydney, Australia; Children's Hospital at Westmead, Sydney, Australia
| | | |
Collapse
|
29
|
Wysocki J, Malecka I, Stryczynska-Kazubska J, Rampakakis E, Kuter B, Wolfson LJ. Varicella in Poland: economic burden in children 1-12 years of age in Poland, 2010-2015. BMC Public Health 2018; 18:410. [PMID: 29587714 PMCID: PMC5870512 DOI: 10.1186/s12889-018-5298-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 03/12/2018] [Indexed: 11/21/2022] Open
Abstract
Background The safety and efficacy of live-attenuated varicella zoster virus (VZV) vaccines in preventing varicella and reducing associated morbidity and mortality in real-world have been previously shown. In Poland, VZV vaccination is only mandatory for certain high-risk individuals. Here, we have conducted an evaluation of the clinical and economic burden of varicella in Poland. Methods Multicenter, retrospective chart review of varicella inpatients and outpatients aged 1–12 years with a primary diagnosis between 2010 and 2015. Varicella-related outcomes included the incidence of complications, the proportion of patients reporting healthcare resource utilization (HCRU), and frequency of HCRU. Direct costs were derived from per patient resource use multiplied by unit costs, and indirect costs were calculated as loss of revenue of caregivers reporting work days missed. The overall annual cost of varicella in Poland was estimated based on the calculated direct and indirect costs per case and the estimated number of varicella cases. All costs are presented in 2015 Polish złoty (PLN) / Euros (€). Results A total of 150 children with varicella were included, of which 75 were outpatients and 75 were inpatients with a mean (± SD) age of 3.9 (±2.6) and 4.2 (±2.3) years, respectively. Complications were experienced by 14.7% of outpatients and 82.7% of inpatients, of which the most common were skin and soft tissue infections and dehydration. The rate of HCRU was as follows: over-the-counter medications (80.0% outpatients, 81.3% inpatients), prescription medications (80.0% outpatients, 93.3% inpatients), tests/procedures (0.0% outpatients, 69.3% inpatients), and allied health professional consults (0.0% outpatients, 24.0% inpatients). Total (direct and indirect) cost per varicella case was 5013.3 PLN (€ 1198.1) for inpatients and 1027.2 PLN (€ 245.5) for outpatients, resulting in an estimated overall annual (2015) cost of varicella in Poland of 178,198,320 PLN (€ 42,588,385) among children aged 1–15 years. Conclusions Significant clinical and economic burden is associated with varicella in Poland. These results may be used to foster discussion related to the implications of implementing routine VZV vaccination in Poland.
Collapse
Affiliation(s)
- Jacek Wysocki
- Poznan University of Medical Sciences, Collegium Maius, Fredry 10, 61-701, Poznań, Poland
| | - Ilona Malecka
- Poznan University of Medical Sciences, Collegium Maius, Fredry 10, 61-701, Poznań, Poland
| | | | | | - Barbara Kuter
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - Lara J Wolfson
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA.
| |
Collapse
|
30
|
Smith DE. The process of popularization – rewriting medical research papers for the layman: Discussion paper. J R Soc Med 2018; 80:634-6. [PMID: 3694602 PMCID: PMC1291052 DOI: 10.1177/014107688708001013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- D E Smith
- Department of English Language Research, University of Birmingham
| |
Collapse
|
31
|
Warren-Gash C, Forbes H, Breuer J. Varicella and herpes zoster vaccine development: lessons learned. Expert Rev Vaccines 2017; 16:1191-1201. [PMID: 29047317 PMCID: PMC5942150 DOI: 10.1080/14760584.2017.1394843] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 10/17/2017] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Before vaccination, varicella zoster virus (VZV), which is endemic worldwide, led to almost universal infection. This neurotropic virus persists lifelong by establishing latency in sensory ganglia, where its reactivation is controlled by VZV-specific T-cell immunity. Lifetime risk of VZV reactivation (zoster) is around 30%. Vaccine development was galvanised by the economic and societal burden of VZV, including debilitating zoster complications that largely affect older individuals. Areas covered: We describe the story of development, licensing and implementation of live attenuated vaccines against varicella and zoster. We consider the complex backdrop of VZV virology, pathogenesis and immune responses in the absence of suitable animal models and examine the changing epidemiology of VZV disease. We review the vaccines' efficacy, safety, effectiveness and coverage using evidence from trials, observational studies from large routine health datasets and clinical post-marketing surveillance studies and outline newer developments in subunit and inactivated vaccines. Expert commentary: Safe and effective, varicella and zoster vaccines have already made major inroads into reducing the burden of VZV disease globally. As these live vaccines have the potential to reactivate and cause clinical disease, developing alternatives that do not establish latency is an attractive prospect but will require better understanding of latency mechanisms.
Collapse
Affiliation(s)
- Charlotte Warren-Gash
- Faculty of Epidemiology & Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Harriet Forbes
- Faculty of Epidemiology & Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Judith Breuer
- Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
32
|
Meszner Z, Molnar Z, Rampakakis E, Yang HK, Kuter BJ, Wolfson LJ. Economic burden of varicella in children 1-12 Years of age in Hungary, 2011-2015. BMC Infect Dis 2017; 17:495. [PMID: 28705150 PMCID: PMC5513371 DOI: 10.1186/s12879-017-2575-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 06/29/2017] [Indexed: 11/16/2022] Open
Abstract
Background Although live-attenuated varicella-zoster virus (VZV) vaccines have been proven to be safe and effective in preventing varicella and real-word evidence shows routine childhood immunization programs are effective in dramatically reducing varicella associated morbidity and mortality, varicella vaccine is not included in the National Immunization Program (NIP) in Hungary. The purpose of this study was to evaluate the clinical and economic burden associated with varicella in Hungary. Methods This was a multicenter, retrospective, chart review study of patients aged 1–12 years with a primary varicella diagnosis between 2011 and 2015. Healthcare resource utilization (HCRU) associated with varicella, unit costs, and work loss were used to estimate direct and indirect costs. All costs are presented in 2015 HUF / Euros (€). Results 156 children with varicella were included (75 outpatients, 81 inpatients), with a mean age of 4.4 (SD: 2.0) and 3.7 (SD: 2.1) years, respectively. One or more complications were reported by 12.0% of outpatients and 92.6% of inpatients, the most common being dehydration, skin and soft tissue infections, pneumonia, keratoconjunctivitis, and cerebellitis. HCRU estimates included use of over-the-counter (OTC) medications (96.0% outpatients, 53.1% inpatients), prescription medications (9.3% outpatients, 70.4% inpatients), tests/procedures (4.0% outpatients, 97.5% inpatients), and consultation with allied health professionals (2.7% outpatients, 30.9% inpatients). The average duration of hospital stay (inpatients) was 3.6 (95% CI: 3.2, 4.1) days. The total combined direct and indirect cost per varicella case was 228,146.7 Hungarian Forint (HUF)/€ 736.0 for inpatients and 49,790.6 HUF/€ 106.6 for outpatients. The overall annual cost of varicella in Hungary for children aged <15 years in 2015 was estimated at 1,903,332,524.3 HUF/ € 6,139,980.4. Conclusion Varicella is associated with substantial clinical burden in Hungary, resulting in the utilization of a significant amount of healthcare resources. These results support the need for routine vaccination of all healthy children to reduce the varicella-associated disease burden.
Collapse
Affiliation(s)
- Z Meszner
- St. László Hospital for Infectious Diseases, National Institute of Child Health, Budapest, Hungary
| | - Z Molnar
- National Center for Epidemiology, Budapest, Hungary
| | | | - H K Yang
- Merck & Co, Inc., Center for Observational and Real-World Evidence (CORE), MAILSTOP WP97-A243, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - B J Kuter
- Merck & Co, Inc., Center for Observational and Real-World Evidence (CORE), MAILSTOP WP97-A243, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Lara J Wolfson
- Merck & Co, Inc., Center for Observational and Real-World Evidence (CORE), MAILSTOP WP97-A243, 770 Sumneytown Pike, West Point, PA, 19486, USA.
| |
Collapse
|
33
|
Gershon AA. Is chickenpox so bad, what do we know about immunity to varicella zoster virus, and what does it tell us about the future? J Infect 2017; 74 Suppl 1:S27-S33. [PMID: 28646959 PMCID: PMC5726865 DOI: 10.1016/s0163-4453(17)30188-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Varicella and zoster continue to cause significant morbidity and even mortality in children and adults. Complications include bacterial superinfection, central nervous system manifestations such as meningitis, encephalitis, and cerebellar ataxia, and pain syndromes especially post herpetic neuralgia. Many developed countries but not all, are now administering live attenuated varicella vaccine routinely, with a decrease in the incidence of disease, providing personal and herd immunity. There is some controversy, however, in some countries concerning whether a decrease in the circulation of wild type virus will result in loss of immunity to VZV in persons who have already had varicella. This manuscript reviews the complications of varicella and zoster in detail, the reasons for development of vaccines against these diseases, complications of vaccinations, and mechanisms by which immunity to this virus develops and is maintained. There are strong indications that the best way to control disease and spread of this virus is by vaccination against both.
Collapse
Affiliation(s)
- Anne A Gershon
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
34
|
Su JR, Leroy Z, Lewis PW, Haber P, Marin M, Leung J, Jane Woo E, Shimabukuro TT. Safety of Second-Dose Single-Antigen Varicella Vaccine. Pediatrics 2017; 139:peds.2016-2536. [PMID: 28174201 PMCID: PMC6434520 DOI: 10.1542/peds.2016-2536] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/22/2016] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVE In 2006, routine 2-dose varicella vaccination for children was recommended to improve control of varicella. We assessed the safety of second-dose varicella vaccination. METHODS We identified second-dose single-antigen varicella vaccine reports in the Vaccine Adverse Event Reporting System during 2006 to 2014 among children aged 4 to 18 years. We analyzed reports by age group (4-6 and 7-18 years), sex, serious or nonserious status, most common adverse events (AEs), and whether other vaccines were administered concomitantly with varicella vaccine. We reviewed serious reports of selected AEs and conducted empirical Bayesian data mining to detect disproportional reporting of AEs. RESULTS We identified 14 641 Vaccine Adverse Event Reporting System reports after second-dose varicella vaccination, with 494 (3%) classified as serious. Among nonserious reports, injection site reactions were most common (48% of children aged 4-6 years, 38% of children aged 7-18 years). The most common AEs among serious reports were pyrexia (31%) for children aged 4 to 6 years and headache (28%) and vomiting (27%) for children aged 7 to 18 years. Serious reports of selected AEs included anaphylaxis (83), meningitis (5), encephalitis (16), cellulitis (52), varicella (6), herpes zoster (6), and deaths (7). One immunosuppressed adolescent was reported with vaccine-strain herpes zoster. Only previously known AEs were reported more frequently after second-dose varicella vaccination compared with other vaccines. CONCLUSIONS We identified no new or unexpected safety concerns for second-dose varicella vaccination. Robust safety monitoring remains an important component of the national varicella vaccination program.
Collapse
Affiliation(s)
- John R. Su
- Immunization Safety Office, Division of Healthcare Quality Promotion, National Center for Emerging and Zoonotic Infectious Disease
| | - Zanie Leroy
- School Health Branch, Division of Population Health, National Center for Chronic Disease Prevention and Health Promotion
| | - Paige W. Lewis
- Immunization Safety Office, Division of Healthcare Quality Promotion, National Center for Emerging and Zoonotic Infectious Disease
| | - Penina Haber
- Immunization Safety Office, Division of Healthcare Quality Promotion, National Center for Emerging and Zoonotic Infectious Disease
| | - Mona Marin
- Epidemiology Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jessica Leung
- Epidemiology Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Emily Jane Woo
- Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Springs, MD
| | - Tom T. Shimabukuro
- Immunization Safety Office, Division of Healthcare Quality Promotion, National Center for Emerging and Zoonotic Infectious Disease
| |
Collapse
|
35
|
Marin M, Marti M, Kambhampati A, Jeram SM, Seward JF. Global Varicella Vaccine Effectiveness: A Meta-analysis. Pediatrics 2016; 137:e20153741. [PMID: 26908671 DOI: 10.1542/peds.2015-3741] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2015] [Indexed: 11/24/2022] Open
Abstract
CONTEXT Several varicella vaccines are available worldwide. Countries with a varicella vaccination program use 1- or 2-dose schedules. OBJECTIVE We examined postlicensure estimates of varicella vaccine effectiveness (VE) among healthy children. DATA SOURCES Systematic review and descriptive and meta-analysis of Medline, Embase, Cochrane libraries, and CINAHL databases for reports published during 1995-2014. STUDY SELECTION Publications that reported original data on dose-specific varicella VE among immunocompetent children. DATA EXTRACTION We used random effects meta-analysis models to obtain pooled one dose VE estimates by disease severity (all varicella and moderate/severe varicella). Within each severity category, we assessed pooled VE by vaccine and by study design. We used descriptive statistics to summarize 1-dose VE against severe disease. For 2-dose VE, we calculated pooled estimates against all varicella and by study design. RESULTS The pooled 1-dose VE was 81% (95% confidence interval [CI]: 78%-84%) against all varicella and 98% (95% CI: 97%-99%) against moderate/severe varicella with no significant association between VE and vaccine type or study design (P > .1). For 1 dose, median VE for prevention of severe disease was 100% (mean = 99.4%). The pooled 2-dose VE against all varicella was 92% (95% CI: 88%-95%), with similar estimates by study design. LIMITATIONS VE was assessed primarily during outbreak investigations and using clinically diagnosed varicella. CONCLUSIONS One dose of varicella vaccine was moderately effective in preventing all varicella and highly effective in preventing moderate/severe varicella, with no differences by vaccine. The second dose adds improved protection against all varicella.
Collapse
Affiliation(s)
- Mona Marin
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia; and
| | - Melanie Marti
- Department of Immunization, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland
| | - Anita Kambhampati
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia; and
| | - Stanley M Jeram
- Department of Immunization, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland
| | - Jane F Seward
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia; and
| |
Collapse
|
36
|
Gershon AA, Breuer J, Cohen JI, Cohrs RJ, Gershon MD, Gilden D, Grose C, Hambleton S, Kennedy PGE, Oxman MN, Seward JF, Yamanishi K. Varicella zoster virus infection. Nat Rev Dis Primers 2015; 1:15016. [PMID: 27188665 PMCID: PMC5381807 DOI: 10.1038/nrdp.2015.16] [Citation(s) in RCA: 402] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Infection with varicella zoster virus (VZV) causes varicella (chickenpox), which can be severe in immunocompromised individuals, infants and adults. Primary infection is followed by latency in ganglionic neurons. During this period, no virus particles are produced and no obvious neuronal damage occurs. Reactivation of the virus leads to virus replication, which causes zoster (shingles) in tissues innervated by the involved neurons, inflammation and cell death - a process that can lead to persistent radicular pain (postherpetic neuralgia). The pathogenesis of postherpetic neuralgia is unknown and it is difficult to treat. Furthermore, other zoster complications can develop, including myelitis, cranial nerve palsies, meningitis, stroke (vasculopathy), retinitis, and gastroenterological infections such as ulcers, pancreatitis and hepatitis. VZV is the only human herpesvirus for which highly effective vaccines are available. After varicella or vaccination, both wild-type and vaccine-type VZV establish latency, and long-term immunity to varicella develops. However, immunity does not protect against reactivation. Thus, two vaccines are used: one to prevent varicella and one to prevent zoster. In this Primer we discuss the pathogenesis, diagnosis, treatment, and prevention of VZV infections, with an emphasis on the molecular events that regulate these diseases. For an illustrated summary of this Primer, visit: http://go.nature.com/14xVI1.
Collapse
Affiliation(s)
- Anne A Gershon
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, New York 10032, USA
| | - Judith Breuer
- Department of Infection and Immunity, University College London, UK
| | - Jeffrey I Cohen
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Massachusetts, USA
| | - Randall J Cohrs
- Departments of Neurology and Microbiology and Immunology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Michael D Gershon
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Don Gilden
- Departments of Neurology and Microbiology and Immunology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Charles Grose
- Division of Infectious Diseases/Virology, Children's Hospital, University of Iowa, Iowa City, Iowa, USA
| | - Sophie Hambleton
- Primary Immunodeficiency Group, Institute of Cellular Medicine, Newcastle University Medical School, Newcastle upon Tyne, UK
| | - Peter G E Kennedy
- Department of Neurology, Institute of Neurological Sciences, Southern General Hospital, Glasgow University, Glasgow, Scotland, UK
| | - Michael N Oxman
- Infectious Diseases Section, Medicine Service, Veterans Affairs San Diego Healthcare System, Division of Infectious Diseases, Department of Medicine, University of California San Diego School of Medicine, San Diego, California, USA
| | - Jane F Seward
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Koichi Yamanishi
- Research Foundation for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
37
|
Vacunas frente al virus de la varicela zóster. Enferm Infecc Microbiol Clin 2015; 33:411-23. [DOI: 10.1016/j.eimc.2015.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 05/20/2015] [Indexed: 01/28/2023]
|
38
|
De Gregorio E, Rappuoli R. From empiricism to rational design: a personal perspective of the evolution of vaccine development. Nat Rev Immunol 2014; 14:505-14. [PMID: 24925139 PMCID: PMC7096907 DOI: 10.1038/nri3694] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Vaccination, which is the most effective medical intervention that has ever been introduced, originated from the observation that individuals who survived a plague or smallpox would not get the disease twice. To mimic the protective effects of natural infection, Jenner - and later Pasteur - inoculated individuals with attenuated or killed disease-causing agents. This empirical approach inspired a century of vaccine development and the effective prophylaxis of many infectious diseases. From the 1980s, several waves of new technologies have enabled the development of novel vaccines that would not have been possible using the empirical approach. The technological revolution in the field of vaccination is now continuing, and it is delivering novel and safer vaccines. In this Timeline article, we provide our views on the transition from empiricism to rational vaccine design.
Collapse
Affiliation(s)
| | - Rino Rappuoli
- Novartis Vaccines, Via Fiorentina 1, Siena, 53100 Italy
| |
Collapse
|
39
|
Macartney K, Heywood A, McIntyre P. Vaccines for post-exposure prophylaxis against varicella (chickenpox) in children and adults. Cochrane Database Syst Rev 2014; 2014:CD001833. [PMID: 24954057 PMCID: PMC7061782 DOI: 10.1002/14651858.cd001833.pub3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND The prevention of varicella (chickenpox) using live attenuated varicella vaccines has been demonstrated both in randomised controlled trials (RCTs) and in population-based immunisation programmes in countries such as the United States and Australia. Many countries do not routinely immunise children against varicella and exposures continue to occur. Although the disease is often mild, complications such as secondary bacterial infection, pneumonitis and encephalitis occur in about 1% of cases, usually leading to hospitalisation. The use of varicella vaccine in persons who have recently been exposed to the varicella zoster virus has been studied as a form of post-exposure prophylaxis (PEP). OBJECTIVES To assess the efficacy and safety of vaccines for use as PEP for the prevention of varicella in children and adults. SEARCH METHODS We searched CENTRAL (2014, Issue 1), MEDLINE (1966 to March week 1, 2014), EMBASE (January 1990 to March 2014) and LILACS (1982 to March 2014). We searched for unpublished trials registered on the clinicaltrials.gov and WHO ICTRP websites. SELECTION CRITERIA RCTs and quasi-RCTs of varicella vaccine for PEP compared with placebo or no intervention. The outcome measures were efficacy in prevention of clinical cases and/or laboratory-confirmed clinical cases and adverse events following vaccination. DATA COLLECTION AND ANALYSIS Two review authors independently extracted and analysed data using Review Manager software. MAIN RESULTS We identified three trials involving 110 healthy children who were siblings of household contacts. The included trials varied in study quality, vaccine used, length of follow-up and outcomes measured and, as such, were not suitable for meta-analysis. We identified high or unclear risk of bias in two of the three included studies. Overall, 13 out of 56 vaccine recipients (23%) developed varicella compared with 42 out of 54 placebo (or no vaccine) recipients (78%). Of the vaccine recipients who developed varicella, the majority only had mild disease (with fewer than 50 skin lesions). In the three trials, most participants received PEP within three days following exposure; too few participants were vaccinated four to five days post-exposure to ascertain the efficacy of vaccine given more than three days after exposure. No included trial reported on adverse events following immunisation. AUTHORS' CONCLUSIONS These small trials suggest varicella vaccine administered within three days to children following household contact with a varicella case reduces infection rates and severity of cases. We identified no RCTs for adolescents or adults. Safety was not adequately addressed.
Collapse
Affiliation(s)
- Kristine Macartney
- Children's Hospital at Westmead and University of SydneyNational Centre for Immunisation Research and Surveillance of Vaccine Preventable DiseasesLocked Bag 4001WestmeadSydneyNSWAustralia2145
| | - Anita Heywood
- University of New South WalesSchool of Public Health and Community MedicineLevel 2, Samuels BuildingGate 11, Botany StreetKensingtonNSWAustralia2052
| | - Peter McIntyre
- Children's Hospital at Westmead and University of SydneyNational Centre for Immunisation Research and Surveillance of Vaccine Preventable DiseasesLocked Bag 4001WestmeadSydneyNSWAustralia2145
| | | |
Collapse
|
40
|
Affiliation(s)
- Kristine Macartney
- National Centre for Immunisation Research and Surveillance, Kids Research Institute, The Children's Hospital at Westmead, Westmead, NSW 2145, Australia.
| |
Collapse
|
41
|
Zerboni L, Sen N, Oliver SL, Arvin AM. Molecular mechanisms of varicella zoster virus pathogenesis. Nat Rev Microbiol 2014; 12:197-210. [PMID: 24509782 PMCID: PMC4066823 DOI: 10.1038/nrmicro3215] [Citation(s) in RCA: 281] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Varicella zoster virus (VZV) is the causative agent of varicella (chickenpox) and zoster (shingles). Investigating VZV pathogenesis is challenging as VZV is a human-specific virus and infection does not occur, or is highly restricted, in other species. However, the use of human tissue xenografts in mice with severe combined immunodeficiency (SCID) enables the analysis of VZV infection in differentiated human cells in their typical tissue microenvironment. Xenografts of human skin, dorsal root ganglia or foetal thymus that contains T cells can be infected with mutant viruses or in the presence of inhibitors of viral or cellular functions to assess the molecular mechanisms of VZV-host interactions. In this Review, we discuss how these models have improved our understanding of VZV pathogenesis.
Collapse
Affiliation(s)
- Leigh Zerboni
- Departments of Pediatrics and of Microbiology & Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Nandini Sen
- Departments of Pediatrics and of Microbiology & Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Stefan L Oliver
- Departments of Pediatrics and of Microbiology & Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Ann M Arvin
- Departments of Pediatrics and of Microbiology & Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
42
|
Watson B. Varicella-zoster vaccine in the USA: success for control of disease severity, but what next? Expert Rev Anti Infect Ther 2014; 3:105-15. [PMID: 15757461 DOI: 10.1586/14787210.3.1.105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In the period from 1990 to 1994, before the introduction of a varicella vaccine to the USA, approximately 100 deaths in otherwise healthy individuals, children and adolescents under 20 years of age, were attributable to varicella complications. The administration of a single-dose vaccine has now been widespread in the USA for nearly 10 years; however, since the effectiveness of a single dose in children under 13 years of age in an outbreak situation is approximately 80%, consideration of a second booster dose is in progress although not yet recommended. Licensure of a measles-mumps-rubella-varicella vaccine may hasten the recommendation.
Collapse
Affiliation(s)
- Barbara Watson
- Jefferson Medical College, Medical Specialist, Immunization Program, Division of Disease Control, The Philadelphia Department of Public Health, 500S Broad Street, Philadelphia, PA 19146, USA.
| |
Collapse
|
43
|
Davis MM. Successes and remaining challenges after 10 years of varicella vaccination in the USA. Expert Rev Vaccines 2014; 5:295-302. [PMID: 16608428 DOI: 10.1586/14760584.5.2.295] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Beginning in 1995, universal varicella vaccination was recommended in the USA for all children aged 12-18 months, and all susceptible adolescents and adults. Many physicians were initially sceptical about the need to prevent primary varicella. However, with passage of state daycare and school entry mandates for varicella immunization, national varicella vaccination rates increased to approximately 90% by 2004. Several studies have demonstrated concomitant reductions in varicella-related healthcare utilization, costs and varicella-related mortality among children in the vaccinated age group, as well as adults. Remaining challenges include: first, outbreaks of 'breakthrough' varicella in vaccinated populations, which may prompt a second-dose recommendation, and second, possible increases in the incidence of secondary varicella (zoster) among adults whose natural immunity may wane in the absence of endemic varicella. The latter concern highlights the importance of a promising new varicella vaccine for older adults that may be licensed and recommended in the next 2 years.
Collapse
Affiliation(s)
- Matthew M Davis
- University of Michigan, Gerald R Ford School of Public Policy, 300 NIB, 6C23 Ann Arbor, MI 48109-0456, USA.
| |
Collapse
|
44
|
Issa NC, Marty FM, Leblebjian H, Galar A, Shea MM, Antin JH, Soiffer RJ, Baden LR. Live attenuated varicella-zoster vaccine in hematopoietic stem cell transplantation recipients. Biol Blood Marrow Transplant 2013; 20:285-7. [PMID: 24269706 DOI: 10.1016/j.bbmt.2013.11.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 11/18/2013] [Indexed: 01/17/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT) recipients are at risk for varicella-zoster virus (VZV) reactivation. Vaccination may help restore VZV immunity; however, the available live attenuated VZV vaccine (Zostavax) is contraindicated in immunocompromised hosts. We report our experience with using a single dose of VZV vaccine in 110 adult autologous and allogeneic HSCT recipients who were about 2 years after transplantation, free of graft-versus-host disease, and not receiving immunosuppression. One hundred eight vaccine recipients (98.2%) had no clinically apparent adverse events with a median follow-up period of 9.5 months (interquartile range, 6 to 16; range, 2 to 28). Two vaccine recipients (1.8%) developed a skin rash (one zoster-like rash with associated pain, one varicella-like) within 42 days post-vaccination that resolved with antiviral therapy. We could not confirm if these rashes were due to vaccine (Oka) or wild-type VZV. No other possible cases of VZV reactivation have occurred with about 1178 months of follow-up. Live attenuated zoster vaccine appears generally safe in this population when vaccinated as noted; the overall vaccination risk needs to be weighed against the risk of wild-type VZV disease in this high-risk population.
Collapse
Affiliation(s)
- Nicolas C Issa
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts.
| | - Francisco M Marty
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Houry Leblebjian
- Department of Pharmacy, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alicia Galar
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Margaret M Shea
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts
| | - Joseph H Antin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Robert J Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Lindsey R Baden
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
45
|
Downes KJ. 35th Annual Pediatric Infectious Diseases Society Awards. J Pediatric Infect Dis Soc 2013; 2:189-97. [PMID: 26619474 DOI: 10.1093/jpids/pit041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
The following is a compilation of the 2013 Pediatric Infectious Diseases Society Awards presented on Monday, May 6, 2013 during the Pediatric Academic Societies meeting in Washington, DC. Award summaries were adapted from submissions provided by the nominating physician.
Collapse
|
46
|
Baxter R, Ray P, Tran TN, Black S, Shinefield HR, Coplan PM, Lewis E, Fireman B, Saddier P. Long-term effectiveness of varicella vaccine: a 14-Year, prospective cohort study. Pediatrics 2013; 131:e1389-96. [PMID: 23545380 DOI: 10.1542/peds.2012-3303] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Varicella vaccine was licensed in the United States in 1995 for individuals ≥12 months of age. A second dose was recommended in the United States in June 2006. Varicella incidence and vaccine effectiveness were assessed in a 14-year prospective study conducted at Kaiser Permanente Northern California. METHODS A total of 7585 children vaccinated with varicella vaccine in their second year of life in 1995 were followed up prospectively for breakthrough varicella and herpes zoster (HZ) through 2009. A total of 2826 of these children received a second dose in 2006-2009. Incidences of varicella and HZ were estimated and compared with prevaccine era rates. RESULTS In this cohort of vaccinated children, the average incidence of varicella was 15.9 per 1000 person-years, nine- to tenfold lower than in the prevaccine era. Vaccine effectiveness at the end of the study period was 90%, with no indication of waning over time. Most cases of varicella were mild and occurred early after vaccination. No child developed varicella after a second dose. HZ cases were mild, and rates were lower in the cohort of vaccinated children than in unvaccinated children during the prevaccine era (relative risk: 0.61 [95% confidence interval: 0.43-0.89]). CONCLUSIONS This study confirmed that varicella vaccine is effective at preventing chicken pox, with no waning noted over a 14-year period. One dose provided excellent protection against moderate to severe disease, and most cases occurred shortly after the cohort was vaccinated. The study data also suggest that varicella vaccination may reduce the risks of HZ in vaccinated children.
Collapse
Affiliation(s)
- Roger Baxter
- Kaiser Permanente Vaccine Study Center, Oakland, CA 94612, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Goldman GS, King PG. Review of the United States universal varicella vaccination program: Herpes zoster incidence rates, cost-effectiveness, and vaccine efficacy based primarily on the Antelope Valley Varicella Active Surveillance Project data. Vaccine 2013; 31:1680-94. [PMID: 22659447 PMCID: PMC3759842 DOI: 10.1016/j.vaccine.2012.05.050] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 05/10/2012] [Accepted: 05/19/2012] [Indexed: 12/31/2022]
Abstract
In a cooperative agreement starting January 1995, prior to the FDA's licensure of the varicella vaccine on March 17, the Centers for Disease Control and Prevention (CDC) funded the Los Angeles Department of Health Services' Antelope Valley Varicella Active Surveillance Project (AV-VASP). Since only varicella case reports were gathered, baseline incidence data for herpes zoster (HZ) or shingles was lacking. Varicella case reports decreased 72%, from 2834 in 1995 to 836 in 2000 at which time approximately 50% of children under 10 years of age had been vaccinated. Starting in 2000, HZ surveillance was added to the project. By 2002, notable increases in HZ incidence rates were reported among both children and adults with a prior history of natural varicella. However, CDC authorities still claimed that no increase in HZ had occurred in any US surveillance site. The basic assumptions inherent to the varicella cost-benefit analysis ignored the significance of exogenous boosting caused by those shedding wild-type VZV. Also ignored was the morbidity associated with even rare serious events following varicella vaccination as well as the morbidity from increasing cases of HZ among adults. Vaccine efficacy declined below 80% in 2001. By 2006, because 20% of vaccinees were experiencing breakthrough varicella and vaccine-induced protection was waning, the CDC recommended a booster dose for children and, in 2007, a shingles vaccination was approved for adults aged 60 years and older. In the prelicensure era, 95% of adults experienced natural chickenpox (usually as children)-these cases were usually benign and resulted in long-term immunity. Varicella vaccination is less effective than the natural immunity that existed in prevaccine communities. Universal varicella vaccination has not proven to be cost-effective as increased HZ morbidity has disproportionately offset cost savings associated with reductions in varicella disease. Universal varicella vaccination has failed to provide long-term protection from VZV disease.
Collapse
Affiliation(s)
- G S Goldman
- Independent Computer Scientist, P.O. Box 847, Pearblossom, CA 93553, United States.
| | | |
Collapse
|
48
|
Cenoz MG, Martínez-Artola V, Guevara M, Ezpeleta C, Barricarte A, Castilla J. Effectiveness of one and two doses of varicella vaccine in preventing laboratory-confirmed cases in children in Navarre, Spain. Hum Vaccin Immunother 2013; 9:1172-6. [PMID: 23324571 DOI: 10.4161/hv.23451] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Varicella vaccine effectiveness was evaluated in a case-control study in Navarre, Spain, in 2010-2012. The cases were 54 children aged 15 months to 10 years with a diagnosis of varicella confirmed by polymerase-chain-reaction. Each case was matched with eight controls by pediatric practice, district of residence and date of birth. The effectiveness was 87% (95% confidence interval: 60% to 97%) for one dose of vaccine and 97% (80% to 100%) for two doses. A single dose was 93% (34% to 100%) effective in the first year, which declined to 61% (95% CI: -64% to 94%) after the third year. In conclusion, varicella vaccine is highly effective in preventing confirmed cases, although this effect declines over time since the first dose. A second dose helps to reestablish very high levels of effectiveness and to reduce the risk of breakthrough varicella.
Collapse
Affiliation(s)
- Manuel García Cenoz
- Instituto de Salud Pública de Navarra; Pamplona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP); Pamplona, Spain
| | | | | | | | | | | |
Collapse
|
49
|
|
50
|
|