201
|
Payne DC, Selvarangan R, Azimi PH, Boom JA, Englund JA, Staat MA, Halasa NB, Weinberg GA, Szilagyi PG, Chappell J, McNeal M, Klein EJ, Sahni LC, Johnston SH, Harrison CJ, Baker CJ, Bernstein DI, Moffatt ME, Tate JE, Mijatovic-Rustempasic S, Esona MD, Wikswo ME, Curns AT, Sulemana I, Bowen MD, Gentsch JR, Parashar UD. Long-term Consistency in Rotavirus Vaccine Protection: RV5 and RV1 Vaccine Effectiveness in US Children, 2012-2013. Clin Infect Dis 2015; 61:1792-9. [PMID: 26449565 DOI: 10.1093/cid/civ872] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/24/2015] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Using a multicenter, active surveillance network from 2 rotavirus seasons (2012 and 2013), we assessed the vaccine effectiveness of RV5 (RotaTeq) and RV1 (Rotarix) rotavirus vaccines in preventing rotavirus gastroenteritis hospitalizations and emergency department (ED) visits for numerous demographic and secular strata. METHODS We enrolled children hospitalized or visiting the ED with acute gastroenteritis (AGE) for the 2012 and 2013 seasons at 7 medical institutions. Stool specimens were tested for rotavirus by enzyme immunoassay and genotyped, and rotavirus vaccination histories were compared for rotavirus-positive cases and rotavirus-negative AGE controls. We calculated the vaccine effectiveness (VE) for preventing rotavirus associated hospitalizations and ED visits for each vaccine, stratified by vaccine dose, season, clinical setting, age, predominant genotype, and ethnicity. RESULTS RV5-specific VE analyses included 2961 subjects, 402 rotavirus cases (14%) and 2559 rotavirus-negative AGE controls. RV1-specific VE analyses included 904 subjects, 100 rotavirus cases (11%), and 804 rotavirus-negative AGE controls. Over the 2 rotavirus seasons, the VE for a complete 3-dose vaccination with RV5 was 80% (confidence interval [CI], 74%-84%), and VE for a complete 2-dose vaccination with RV1 was 80% (CI, 68%-88%).Statistically significant VE was observed for each year of life for which sufficient data allowed analysis (7 years for RV5 and 3 years for RV1). Both vaccines provided statistically significant genotype-specific protection against predominant circulating rotavirus strains. CONCLUSIONS In this large, geographically and demographically diverse sample of US children, we observed that RV5 and RV1 rotavirus vaccines each provided a lasting and broadly heterologous protection against rotavirus gastroenteritis.
Collapse
Affiliation(s)
- Daniel C Payne
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | | | - Julie A Boom
- Texas Children's Hospital Baylor College of Medicine, Houston, Texas
| | | | | | | | | | - Peter G Szilagyi
- University of Rochester School of Medicine and Dentistry, New York University of California, Los Angeles
| | - James Chappell
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | | | | | | | | | | | | | - Jacqueline E Tate
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Slavica Mijatovic-Rustempasic
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Mathew D Esona
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Mary E Wikswo
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Aaron T Curns
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Iddrisu Sulemana
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Michael D Bowen
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jon R Gentsch
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Umesh D Parashar
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
202
|
Scully IL, Swanson K, Green L, Jansen KU, Anderson AS. Anti-infective vaccination in the 21st century—new horizons for personal and public health. Curr Opin Microbiol 2015; 27:96-102. [DOI: 10.1016/j.mib.2015.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/10/2015] [Indexed: 12/17/2022]
|
203
|
Karmann A, Jurack A, Lukas D. Recommendation of rotavirus vaccination and herd effect: a budget impact analysis based on German health insurance data. THE EUROPEAN JOURNAL OF HEALTH ECONOMICS : HEPAC : HEALTH ECONOMICS IN PREVENTION AND CARE 2015; 16:719-31. [PMID: 25135769 PMCID: PMC4534507 DOI: 10.1007/s10198-014-0624-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 08/05/2014] [Indexed: 05/25/2023]
Abstract
The objective of this study was to assess the budget impact and health effects of introducing rotavirus (RV) vaccination in Saxony, Germany, from a health insurance perspective. Special emphasis is given to the herd effect. We analyzed direct medical and non-medical costs of RV infection for Social Health Insurance between 2007 and 2010 based on 360,000 routine data observations from the AOK PLUS for children below 5 years of age. We compared the actual annual number of RV cases (vaccination scenario) with the number derived from 2005 (no vaccination, base case scenario). The vaccination coverage rate has increased from 5% to 61% between 2007 and 2010. The number of RV cases decreased by 21% from 32,274 in 2007 to 25,614 in 2010. Based on vaccination coverage, the total cost savings per 1,000 children due to RV vaccination was estimated to be 39,686 Euros. The overall share of outpatient costs was 60%. Mean gross cost savings were expected to be 304 Euros per avoided case. The net cost savings were expected to be 19 Euros per avoided case. About 59% of total savings was due to herd protection resulting from increasing vaccine rates. The herd effect per avoided case increased with increasing vaccine coverage. Incidence of RV cases, vaccination costs and days absent from work were sensitive parameters. This retrospective analysis showed that the increase in RV vaccination coverage in Saxony has been budget neutral if not cost saving for sick funds.
Collapse
Affiliation(s)
- Alexander Karmann
- Fakultät Wirtschaftswissenschaften, Gesundheitsökonomisches Zentrum, TU Dresden, 01062 Dresden, Germany
| | - Andrea Jurack
- Lehrstuhl für Allokationstheorie, Fakultät Wirtschaftswissenschaften, TU Dresden, 01062 Dresden, Germany
| | - Daniel Lukas
- Lehrstuhl für Wirtschaftsforschung und Wirtschaftspolitik, Fakultät Wirtschaftswissenschaften, TU Dresden, 01062 Dresden, Germany
| |
Collapse
|
204
|
Abstract
Rotavirus infection is the most common cause of severe diarrhea disease in infants and young children worldwide. Vaccination is the only control measure likely to have a significant impact on the incidence of severe disease. Rotavirus vaccines have reduced the burden of disease in the United States and Europe and vaccine programs are being introduced in Asia and Africa where it is hoped that vaccine will have significant impact on severe infection. Long-term monitoring and strain surveillance are needed to assess the effects of rotavirus immunization programs and to determine whether changes in strain ecology will affect rotavirus vaccine effectiveness.
Collapse
Affiliation(s)
- Penelope H Dennehy
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Hasbro Children's Hospital, 593 Eddy Street, Providence, RI 02903, USA.
| |
Collapse
|
205
|
Zeller M, Donato C, Trovão NS, Cowley D, Heylen E, Donker NC, McAllen JK, Akopov A, Kirkness EF, Lemey P, Van Ranst M, Matthijnssens J, Kirkwood CD. Genome-Wide Evolutionary Analyses of G1P[8] Strains Isolated Before and After Rotavirus Vaccine Introduction. Genome Biol Evol 2015; 7:2473-83. [PMID: 26254487 PMCID: PMC4607516 DOI: 10.1093/gbe/evv157] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Rotaviruses are the most important etiological agent of acute gastroenteritis in young children worldwide. Among the first countries to introduce rotavirus vaccines into their national immunization programs were Belgium (November 2006) and Australia (July 2007). Surveillance programs in Belgium (since 1999) and Australia (since 1989) offer the opportunity to perform a detailed comparison of rotavirus strains circulating pre- and postvaccine introduction. G1P[8] rotaviruses are the most prominent genotype in humans, and a total of 157 G1P[8] rotaviruses isolated between 1999 and 2011 were selected from Belgium and Australia and their complete genomes were sequenced. Phylogenetic analysis showed evidence of frequent reassortment among Belgian and Australian G1P[8] rotaviruses. Although many different phylogenetic subclusters were present before and after vaccine introduction, some unique clusters were only identified after vaccine introduction, which could be due to natural fluctuation or the first signs of vaccine-driven evolution. The times to the most recent common ancestors for the Belgian and Australian G1P[8] rotaviruses ranged from 1846 to 1955 depending on the gene segment, with VP7 and NSP4 resulting in the most recent estimates. We found no evidence that rotavirus population size was affected after vaccine introduction and only six amino acid sites in VP2, VP3, VP7, and NSP1 were identified to be under positive selective pressure. Continued surveillance of G1P[8] strains is needed to determine long-term effects of vaccine introductions, particularly now rotavirus vaccines are implemented in the national immunization programs of an increasing number of countries worldwide.
Collapse
Affiliation(s)
- Mark Zeller
- Laboratory of Clinical Virology, University of Leuven, Leuven, Belgium
| | - Celeste Donato
- Enteric Virus Research Group, Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, VIC, Australia Department of Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Nídia Sequeira Trovão
- Laboratory Evolutionary and Computational Virology, University of Leuven, Leuven, Belgium
| | - Daniel Cowley
- Enteric Virus Research Group, Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | - Elisabeth Heylen
- Laboratory of Clinical Virology, University of Leuven, Leuven, Belgium
| | - Nicole C Donker
- Enteric Virus Research Group, Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | | | - Asmik Akopov
- Laboratory Evolutionary and Computational Virology, University of Leuven, Leuven, Belgium
| | | | - Philippe Lemey
- Laboratory Evolutionary and Computational Virology, University of Leuven, Leuven, Belgium
| | - Marc Van Ranst
- Laboratory of Clinical Virology, University of Leuven, Leuven, Belgium
| | | | - Carl D Kirkwood
- Enteric Virus Research Group, Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, VIC, Australia Department of Microbiology, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
206
|
Rotavirus vaccines: a story of success. Clin Microbiol Infect 2015; 21:735-43. [DOI: 10.1016/j.cmi.2015.01.027] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/28/2015] [Accepted: 01/29/2015] [Indexed: 11/19/2022]
|
207
|
Esona MD, Gautam R, Tam KI, Williams A, Mijatovic-Rustempasic S, Bowen MD. Multiplexed one-step RT-PCR VP7 and VP4 genotyping assays for rotaviruses using updated primers. J Virol Methods 2015; 223:96-104. [PMID: 26231786 DOI: 10.1016/j.jviromet.2015.07.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/23/2015] [Accepted: 07/24/2015] [Indexed: 12/25/2022]
Abstract
The current two-step VP7 and VP4 genotyping RT-PCR assays for rotaviruses have been linked consistently to genotyping failure in an estimated 30% of RVA positive samples worldwide. We have developed a VP7 and VP4 multiplexed one-step genotyping assays using updated primers generated from contemporary VP7 and VP4 sequences. To determine assay specificity and sensitivity, 17 reference virus strains, 6 non-target gastroenteritis viruses and 725 clinical samples carrying the most common VP7 (G1, G2, G3, G4, G9, and G12) and VP4 (P[4], P[6], P[8], P[9] and P[10]) genotypes were tested in this study. All reference RVA strain targets yielded amplicons of the expected sizes and non-target genotypes and gastroenteritis viruses were not detected by either assay. Out of the 725 clinical samples tested, the VP7 and VP4 assays were able to assigned specific genotypes to 711 (98.1%) and 714 (98.5%), respectively. The remaining unassigned samples were re-tested for RVA antigen using EIA and qRT-PCR assays and all were found to be negative. The overall specificity, sensitivity and limit of detection of the VP7 assay were in the ranges of 99.0-100%, 94.0-100% and 8.6×10(1) to 8.6×10(2) copies of RNA/reaction, respectively. For the VP4 assay, the overall specificity, sensitivity and limit of detection assay were in the ranges of 100%, 94.0-100% and ≤1 to 8.6×10(2) copies of RNA/reaction, respectively. Here we report two highly robust, accurate, efficient, affordable and documentable gel-based genotyping systems which are capable of genotyping 97.8% of the six common VP7 and 98.3% of the five common VP4 genotypes of RVA strains which are responsible for approximately 88.2% of all RVA infections worldwide.
Collapse
Affiliation(s)
- Mathew D Esona
- Gastroenteritis and Respiratory Viruses Laboratory Branch, Division of Viral Diseases, NCIRD, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA.
| | - Rashi Gautam
- Gastroenteritis and Respiratory Viruses Laboratory Branch, Division of Viral Diseases, NCIRD, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Ka Ian Tam
- Gastroenteritis and Respiratory Viruses Laboratory Branch, Division of Viral Diseases, NCIRD, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | | | - Slavica Mijatovic-Rustempasic
- Gastroenteritis and Respiratory Viruses Laboratory Branch, Division of Viral Diseases, NCIRD, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Michael D Bowen
- Gastroenteritis and Respiratory Viruses Laboratory Branch, Division of Viral Diseases, NCIRD, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| |
Collapse
|
208
|
Bravo L, Chitraka A, Liu A, Choudhury J, Kumar K, Berezo L, Cimafranca L, Chatterjee P, Garg P, Siriwardene P, Bernardo R, Mehta S, Balasubramanian S, Karkada N, Htay Han H. Reactogenicity and safety of the human rotavirus vaccine, Rotarix™ in The Philippines, Sri Lanka, and India: a post-marketing surveillance study. Hum Vaccin Immunother 2015; 10:2276-83. [PMID: 25424932 PMCID: PMC4896787 DOI: 10.4161/hv.29280] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Regulatory bodies in The Philippines, Sri Lanka, and India require post-marketing surveillance to provide additional safety data on Rotarix™ in real-life settings. In such studies conducted in The Philippines (November 2006 to July 2012; NCT00353366), Sri Lanka (November 2008 to August 2009; NCT00779779), and India (August 2009 to April 2010; NCT00938327), 2 doses of Rotarix™ were administered according to the local prescribing information (PI). The occurrence of at least Grade “2”/”3” solicited adverse event (AE) (fever, vomiting, or diarrhea), within 15 days in The Philippines or 8 days in Sri Lanka and India; unsolicited AEs within 31 days and serious adverse events (SAEs) throughout the study were recorded. Of the 1494, 522, and 332 infants enrolled in The Philippines, Sri Lanka, and India, 14.7% 14.9% and 12.7% infants, respectively recorded at least Grade “2”/”3” solicited AEs. The most commonly reported solicited AEs were irritability in The Philippines (32.2% post-Dose-1; 23.5% post-Dose-2) and India (23.0% post-Dose-1; 13.2% post-Dose-2), and fever (18.0% post-Dose-1; 20.2% post-Dose-2) in Sri Lanka. Unsolicited AEs were recorded in 24.5% (The Philippines), 4.8% (Sri Lanka), and 6.9% (India) of infants. Forty-one SAEs were recorded in the Philippines of which 6 (decreased oral intake with increased sleeping time and constipation; pneumonia, urinary tract infection, and intussusception) were considered by the investigators as causally related to vaccination. One vaccine-unrelated SAE occurred in a Sri Lankan infant. All SAEs resolved and the infants recovered. Two doses of Rotarix™, administered to healthy infants according to local PI, were well tolerated in The Philippines, Sri Lanka, and India.
Collapse
Affiliation(s)
- Lulu Bravo
- a National Institutes of Health-University of The Philippines Manila; Manila, The Philippines
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
209
|
European Society for Paediatric Infectious Diseases consensus recommendations for rotavirus vaccination in Europe: update 2014. Pediatr Infect Dis J 2015; 34:635-43. [PMID: 25860532 DOI: 10.1097/inf.0000000000000683] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The first evidence-based recommendations for rotavirus (RV) vaccination in Europe were prepared at the time of licensure of 2 live oral RV vaccines (Rotarix, GlaxoSmithKline Biologicals, and RotaTeq, Sanofi Pasteur MSD) in 2006 and published in 2008. Since then several countries in Europe and more globally have adopted universal RV vaccination of all healthy infants as part of their national immunization programs (NIPs). The experience from these NIPs has produced a wealth of post-introduction effectiveness data that, together with the evidence from prelicensure efficacy trials presented in the 2008 Recommendations, support the case of RV vaccination in Europe. The prelicensure safety trials of Rotarix and RotaTeq, each in populations of more than 60,000 infants, did not reveal risk of intussusception (IS), but postvaccination surveillance in several countries, particularly Australia and Mexico, has established that the risk of IS for both vaccines after the first dose might be between 1:50,000 and 1:80,000. Although it may be argued that the risk is acceptable vis-à-vis the great benefits of RV vaccination, this argument alone may not suffice, and every effort should be made to reduce the risk of IS. Considerable evidence, including postvaccination surveillance data from Germany, suggests that the risk of IS can be reduced by early administration of the first dose of oral RV vaccine. The previous European Society for Paediatric Infectious Diseases/European Society for Paediatric Gastroenterology, Hepatology and Nutrition recommendations held that the first dose of oral RV vaccine should be given between 6 and 12 weeks of age; this recommendation is sustained but with an emphasis toward the lower range of the recommended age, that is, preferably between 6 and 8 weeks of age. At the time of the earlier recommendations, experience of RV vaccination in premature infants and other special target groups was limited. It is now recommended with greater confidence than before that prematurely born infants should be vaccinated according to their calendar age as recommended for full-term infants. It is now strongly recommended that all HIV-infected or HIV-exposed infants should be vaccinated with oral RV vaccine. Although specific information on many immunodeficiencies is lacking, infants with known severe combined immunodeficiency should not receive live RV vaccine.
Collapse
|
210
|
Evidence of herd immunity and sustained impact of rotavirus vaccination on the reduction of rotavirus-related medical encounters among infants from 2006 through 2011 in the United States. Pediatr Infect Dis J 2015; 34:615-20. [PMID: 25831421 DOI: 10.1097/inf.0000000000000702] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Rotavirus (RV) is the leading cause of severe acute gastroenteritis among young children. Since the US licensure of the pentavalent RV vaccine (RV5) and the monovalent RV vaccine (RV1), a decline of RV activity has been observed. OBJECTIVE To describe patterns of RV-related health care utilization among infants receiving RV vaccines (RVVs). METHODS A large national health insurance claims database was used to identify infants born from January 2002 through July 2011. From this cohort, infants were divided into three groups: (1) those who received a RVV, (2) those receiving a diphtheria, tetanus, and acellular pertussis (DTaP) vaccine before the introduction of RVV (February 2006), and (3) those receiving DTaP without a concurrent RVV during the period of RVV availability. Study outcomes were rotavirus gastroenteritis (RGE) and acute gastroenteritis. Longitudinal, seasonal RGE incidence patterns among the RVV cohort (n = 140,952) were compared with the referent DTaP-vaccine cohort (n = 131,529). RESULTS More than 91% of administered RVV were RV5. Mean peak incidence of RV medical encounters in RV-vaccinated infants was 95-96% lower than among DTaP-vaccinated infants who did not receive RVV. RGE incidence among the non-RV-vaccinated DTaP recipients in the RVV-available period (110 per 100,000 infants) was lower than among DTaP recipients in the pre-RVV period (151 per 100,000 infants). The highest RGE incidence in the 2007-2011 period was among older non-RV-vaccinated infants. CONCLUSIONS Analysis of a national medical claims database indicates a sustained and substantial decrease in the seasonal RV medical claims pattern after the introduction of RVV. This analysis also reveals evidence of herd immunity, although unvaccinated infants continue to be at risk and contribute to smaller seasonal peaks in RV disease activity.
Collapse
|
211
|
Sharma S, Nordgren J. Rotavirus vaccines in developing countries: issues and future considerations. Future Virol 2015. [DOI: 10.2217/fvl.15.32] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Sumit Sharma
- Division of Molecular Virology, Department of Clinical & Experimental Medicine, University of Linköping, 58185 Linköping, Sweden
| | - Johan Nordgren
- Division of Molecular Virology, Department of Clinical & Experimental Medicine, University of Linköping, 58185 Linköping, Sweden
| |
Collapse
|
212
|
Urueña A, Pippo T, Betelu MS, Virgilio F, Hernández L, Giglio N, Gentile Á, Diosque M, Vizzotti C. Cost-effectiveness analysis of rotavirus vaccination in Argentina. Vaccine 2015; 33 Suppl 1:A126-34. [DOI: 10.1016/j.vaccine.2014.12.074] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 12/11/2014] [Accepted: 12/12/2014] [Indexed: 11/30/2022]
|
213
|
Guerra SF, Linhares AC, Mascarenhas JDP, Oliveira A, Justino MCA, Soares LS, Müller EC, Brasil P, Tuboi S, Ortega-Barria E, Colindres R. Rotavirus strain surveillance for three years following the introduction of rotavirus vaccine into Belém, Brazil. J Med Virol 2015; 87:1303-10. [DOI: 10.1002/jmv.24183] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2015] [Indexed: 12/22/2022]
Affiliation(s)
| | | | | | | | | | - Luana S. Soares
- Evandro Chagas Institute, Health Surveillance Secretariat; Belém Brazil
| | | | - Patrícia Brasil
- Clinical Research Institute Evandro Chagas, Oswaldo Cruz Foundation; Rio de Janeiro Brazil
| | | | | | | |
Collapse
|
214
|
Effectiveness and impact of rotavirus vaccines in Europe, 2006–2014. Vaccine 2015; 33:2097-107. [DOI: 10.1016/j.vaccine.2015.03.016] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 03/05/2015] [Accepted: 03/06/2015] [Indexed: 11/17/2022]
|
215
|
Bhandari N, Rongsen-Chandola T, Bavdekar A, John J, Antony K, Taneja S, Goyal N, Kawade A, Kang G, Rathore SS, Juvekar S, Muliyil J, Arya A, Shaikh H, Abraham V, Vrati S, Proschan M, Kohberger R, Thiry G, Glass R, Greenberg HB, Curlin G, Mohan K, Harshavardhan GVJA, Prasad S, Rao TS, Boslego J, Bhan MK. Efficacy of a monovalent human-bovine (116E) rotavirus vaccine in Indian children in the second year of life. Vaccine 2015; 32 Suppl 1:A110-6. [PMID: 25091663 DOI: 10.1016/j.vaccine.2014.04.079] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
UNLABELLED Rotavirus gastroenteritis is one of the leading causes of diarrhea in Indian children less than 2 years of age. The 116E rotavirus strain was developed as part of the Indo-US Vaccine Action Program and has undergone efficacy trials. This paper reports the efficacy and additional safety data in children up to 2 years of age. In a double-blind placebo controlled multicenter trial, 6799 infants aged 6-7 weeks were randomized to receive three doses of an oral human-bovine natural reassortant vaccine (116E) or placebo at ages 6, 10, and 14 weeks. The primary outcome was severe (≥11 on the Vesikari scale) rotavirus gastroenteritis. Efficacy outcomes and adverse events were ascertained through active surveillance. We randomly assigned 4532 and 2267 subjects to receive vaccine and placebo, respectively, with over 96% subjects receiving all three doses of the vaccine or placebo. The per protocol analyses included 4354 subjects in the vaccine and 2187 subjects in the placebo group. The overall incidence of severe RVGE per 100 person years was 1.3 in the vaccine group and 2.9 in the placebo recipients. Vaccine efficacy against severe rotavirus gastroenteritis in children up to 2 years of age was 55.1% (95% CI 39.9 to 66.4; p<0.0001); vaccine efficacy in the second year of life of 48.9% (95% CI 17.4 to 68.4; p=0.0056) was only marginally less than in the first year of life [56.3% (95% CI 36.7 to 69.9; p<0.0001)]. The number of infants needed to be immunized to prevent one episode of severe RVGE in the first 2 years of life was 40 (95% CI 28.0 to 63.0) and for RVGE of any severity, it was 21 (95% CI 16.0 to 32.0). Serious adverse events were observed at the same rates in the two groups. None of the eight intussusception events occurred within 30 days of a vaccine dose and all were reported only after the third dose. The sustained efficacy of the 116E in the second year of life is reassuring. CLINICAL TRIAL REGISTRY The trial is registered with Clinical Trial Registry-India (# CTRI/2010/091/000102) and Clinicaltrials.gov (# NCT01305109).
Collapse
Affiliation(s)
- Nita Bhandari
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | | | | | - Jacob John
- Christian Medical College, Vellore, Tamil Nadu, India
| | | | - Sunita Taneja
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | - Nidhi Goyal
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | - Anand Kawade
- KEM Hospital Research Centre, Pune, Maharashtra, India
| | | | - Sudeep Singh Rathore
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | | | | | - Alok Arya
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | - Hanif Shaikh
- KEM Hospital Research Centre, Pune, Maharashtra, India
| | - Vinod Abraham
- Christian Medical College, Vellore, Tamil Nadu, India
| | - Sudhanshu Vrati
- Translational Health Science and Technology Institute, Gurgaon, Haryana, India; National Institute of Immunology, New Delhi, India
| | | | | | - Georges Thiry
- Advancing Rotavirus Vaccines Development Project, PATH, France
| | - Roger Glass
- National Institutes of Health, Bethesda, MD, USA
| | | | | | - Krishna Mohan
- Bharat Biotech International Limited, Genome Valley, Andhra Pradesh, India
| | | | - Sai Prasad
- Bharat Biotech International Limited, Genome Valley, Andhra Pradesh, India
| | - T S Rao
- Department of Biotechnology, Ministry of Science and Technology, Government of India, India
| | | | | | | |
Collapse
|
216
|
Kompithra RZ, Paul A, Manoharan D, Babji S, Sarkar R, Mathew LG, Kang G. Immunogenicity of a three dose and five dose oral human rotavirus vaccine (RIX4414) schedule in south Indian infants. Vaccine 2015; 32 Suppl 1:A129-33. [PMID: 25091666 DOI: 10.1016/j.vaccine.2014.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIM This study was undertaken to compare the immunogenicity of a three dose and five dose schedule of an oral live-attenuated human rotavirus vaccine, Rotarix in south Indian infants. METHOD Healthy infants (N=90), six to seven weeks of age were enrolled to receive three doses (n=45) or five doses of Rotarix vaccine (n=45) along with other scheduled vaccines, each dose separated by a four week interval. Blood samples were taken before vaccination and one month post-dose three in the Rotarix three dose group and one month post-dose five in the Rotarix five dose group; all were tested for anti-rotavirus IgA by an antibody sandwich enzyme immunoassay. RESULTS At baseline, >50% of infants had >20 units of anti-rotavirus IgA. The seroconversion rates after three and five doses were low and not significantly different in the two groups. However, among vaccine responders, children seropositive at baseline showed a much greater absolute increase in IgA antibody levels than children seronegative at baseline. CONCLUSIONS Rotarix vaccine showed low immunogenicity in south Indian children and increasing the number of doses did not increase the proportion of infants seroconverting after vaccination.
Collapse
Affiliation(s)
| | - Anu Paul
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Divya Manoharan
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Sudhir Babji
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Rajiv Sarkar
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Leni G Mathew
- Department of Paediatrics, Christian Medical College, Vellore, India
| | - Gagandeep Kang
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, India.
| |
Collapse
|
217
|
Kulkarni R, Arora R, Arora R, Chitambar SD. Sequence analysis of VP7 and VP4 genes of G1P[8] rotaviruses circulating among diarrhoeic children in Pune, India: a comparison with Rotarix and RotaTeq vaccine strains. Vaccine 2015; 32 Suppl 1:A75-83. [PMID: 25091685 DOI: 10.1016/j.vaccine.2014.03.080] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND The G1P[8] rotaviruses are a common cause of rotavirus diarrhoea among children in India. Two rotavirus vaccines licensed in India, Rotarix and RotaTeq, contain strains with G1 and P[8] genotypes. A comparative analysis of these genotypes in the live rotavirus vaccines with circulating rotavirus strains is essential for assessment of rotavirus diversity. METHODS G1P[8] strains detected during rotavirus surveillance among diarrhoeic children hospitalized in Pune in 1992-1993 and 2006-2008, were included in the study. Amplification, sequencing and phylogenetic analysis of the VP7 and VP4 genes were carried out for identification of the G1 and P[8] lineages, respectively. Antigenic epitopes of VP7 and VP4 encoded proteins were compared to determine the differences between the G1P[8] strains from Pune and the vaccine strains. RESULTS G1-Lineage 1, P[8]-Lineage 3 strains were predominant in Pune during 1992-1993 and 2006-2008. Strains of G1-Lineage 2, P[8]-Lineage 3 and G1-Lineage 1, P[8]-Lineage 4 were detected at low levels during 2006-2008. The G1-Lineage 1, P[8]-Lineage 3 strains showed up to eight amino acid changes, each in the VP7 and VP4 epitopes, with respect to the Rotarix vaccine strain (G1-Lineage 2, P[8]-Lineage 1) and the G1 (Lineage-3) and P[8] (Lineage 2) components of the RotaTeq vaccine. The G1-Lineage 2 strains were closer to both vaccine strains with no or only two amino acid substitutions in the VP7 epitopes. The divergent P[8]-Lineage 4 (OP354-like) strains showed fourteen and fifteen amino acid differences, with Rotarix and RotaTeq vaccine strains, respectively, in the VP4 epitopes. CONCLUSION The differences between the G1P[8] strains in Pune and the G1 and P[8] components of the vaccine strains need to be described for appropriate evaluation of vaccine shedding. Continuous monitoring of the G1P[8] subgenotypic lineages would be necessary to study any long term impact of vaccine use on G1P[8] strain evolution.
Collapse
Affiliation(s)
- Ruta Kulkarni
- Enteric Viruses Group, National Institute of Virology, 20-A, Dr. Ambedkar Road, Pune-411001, India
| | - Ritu Arora
- Enteric Viruses Group, National Institute of Virology, 20-A, Dr. Ambedkar Road, Pune-411001, India
| | - Rashmi Arora
- Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research, Ansari Nagar, New Delhi-110029, India
| | - Shobha D Chitambar
- Enteric Viruses Group, National Institute of Virology, 20-A, Dr. Ambedkar Road, Pune-411001, India.
| |
Collapse
|
218
|
Paul S, Sahoo J. Four new vaccines for routine immunization in India: what about hemophilus influenza B and pneumococcal vaccine? J Family Med Prim Care 2015; 4:9-12. [PMID: 25810981 PMCID: PMC4367014 DOI: 10.4103/2249-4863.152238] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Immunization is the process whereby a person is made immune or resistant to an infectious disease, typically by the administration of a vaccine. The Expanded Program on Immunization (EPI) was flagged off in India in 1978. According to the recommendation of National technical advisory group of India (NATGI), Government of India is going to include four new vaccines in the UIP for whole India. The four new vaccines are Inactivated Poliomyelitis Vaccine (IPV) for polio, rota viral vaccine, vaccine against rubella, and Japanese encephalitis vaccine (179 districts in India). Here, authors have tried to show a comparative descriptive analysis of the hemophilus influenza and pneumococcal pneumonia with rota virus, so that in near future Government of India can also consider their inclusion in the national UIP. In India, 39.2% of all diarrheal death are due to rota virus, whereas 0.72 million deaths are due to hemophilus influenza B and 1.3 million are due to pneumococcal pneumonia in <5 years age-group. India's indigenous developed rota viral vaccine's (Rotavac) efficacy is 56% in 1(st) year compared to H influenza B (Hib) efficacy 95% and PCV13 vaccine "3 + 1" dose efficacy 100% (South Africa). Rotarix incremental cost-effectiveness ratio is US $21.4 to US $34 per disability-adjusted life years (DALYs) compared to Hib US $ 819 per DALYs in India. In case of pneumococcal vaccine, India needs more trails on the serotype specificity, efficacy, and cost-effectiveness but there is enough evidence that hemophilus influenza burden is high in India and the present Hib vaccine is safe and highly effective. In future with the help of donor agencies, India should include the hemophilus influenza B and pneumococcal pneumonia vaccine in national UIP which will save millions of poor children's life.
Collapse
Affiliation(s)
- Sourabh Paul
- Department of Community Medicine and Family Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Jyotiranjan Sahoo
- Department of Community Medicine and Family Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| |
Collapse
|
219
|
Benhafid M, Elomari N, Azzouzi Idrissi M, Rguig A, Gentsch JR, Parashar U, Elaouad R. Effect of monovalent rotavirus vaccine on rotavirus disease burden and circulating rotavirus strains among children in Morocco. J Med Virol 2015; 87:944-53. [PMID: 25753952 DOI: 10.1002/jmv.24122] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2014] [Indexed: 12/29/2022]
Abstract
Rotarix(TM) vaccine was introduced into the National Program of Immunization of Morocco in October 2010, reaching quickly 87% of the target population of children nationally. The incidence of rotavirus gastroenteritis and the prevalence of circulating rotavirus strains has been monitored in three sentinel hospitals since June 2006. The average percentage of rotavirus positive cases among all children under 5 years old hospitalized for gastroenteritis during the pre-vaccine period (2006-2010) was 44%. This percentage dropped to 29%, 15% and 24% in the 3 years post vaccine introduction (2011, 2012 and 2013), which is a decline of 34%, 66%, and 45%, respectively. Declines in prevalence were greatest among children 0-1 years of age (53%) and were most prominent during the winter and autumn rotavirus season. The prevalence of the G2P[4] and G9P[8] genotype sharply increased in the post vaccine period (2011-2013) compared to the previous seasons (2006-2010). Rotavirus vaccines have reduced greatly the number of children hospitalized due to rotavirus infection at the three sentinel hospitals; it is however unclear if the predominance of G2P[4] and G9P[8] genotypes is related to the vaccine introduction, or if this is attributable to normal genotype fluctuations. Continued surveillance will be pivotal to answer this question in the future.
Collapse
Affiliation(s)
- Mohammed Benhafid
- Virology Laboratory, National Institute of Hygiene, Ministry of Health, Rabat, Morocco
| | | | | | | | | | | | | |
Collapse
|
220
|
Collins P, Mulherin E, O'Shea H, Cashman O, Lennon G, Pidgeon E, Coughlan S, Hall W, Fanning S. Changing patterns of rotavirus strains circulating in Ireland: Re-emergence of G2P[4] and identification of novel genotypes in Ireland. J Med Virol 2015; 87:764-73. [DOI: 10.1002/jmv.24095] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2014] [Indexed: 11/06/2022]
Affiliation(s)
- P.J. Collins
- Department of Biological Sciences; Cork Institute of Technology; Cork Ireland
| | - Emily Mulherin
- UCD-Centre for Food Safety; School of Public Health; Physiotherapy and Population Science; Dublin Ireland
| | - Helen O'Shea
- Department of Biological Sciences; Cork Institute of Technology; Cork Ireland
| | - Olivia Cashman
- Department of Biological Sciences; Cork Institute of Technology; Cork Ireland
| | - Grainne Lennon
- Department of Biological Sciences; Cork Institute of Technology; Cork Ireland
| | - Eugene Pidgeon
- National Virus Reference Laboratory; University College Dublin; Dublin Ireland
| | - Suzie Coughlan
- National Virus Reference Laboratory; University College Dublin; Dublin Ireland
| | - William Hall
- National Virus Reference Laboratory; University College Dublin; Dublin Ireland
| | - Séamus Fanning
- UCD-Centre for Food Safety; School of Public Health; Physiotherapy and Population Science; Dublin Ireland
- School of Veterinary Medicine; University College Dublin; Dublin Ireland
- Institute for Global Food Security; Queen's University Belfast; Belfast Ireland
| |
Collapse
|
221
|
Rotavirus vaccines roll-out in resource-deprived regions. THE LANCET. INFECTIOUS DISEASES 2015; 15:368-70. [PMID: 25638520 DOI: 10.1016/s1473-3099(14)71089-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
222
|
Bilcke J, Chapman R, Atchison C, Cromer D, Johnson H, Willem L, Cox M, Edmunds WJ, Jit M. Quantifying Parameter and Structural Uncertainty of Dynamic Disease Transmission Models Using MCMC: An Application to Rotavirus Vaccination in England and Wales. Med Decis Making 2015; 35:633-47. [PMID: 25623063 DOI: 10.1177/0272989x14566013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 11/23/2014] [Indexed: 11/15/2022]
Abstract
BACKGROUND Two vaccines (Rotarix and RotaTeq) are highly effective at preventing severe rotavirus disease. Rotavirus vaccination has been introduced in the United Kingdom and other countries partly based on modeling and cost-effectiveness results. However, most of these models fail to account for the uncertainty about several vaccine characteristics and the mechanism of vaccine action. METHODS A deterministic dynamic transmission model of rotavirus vaccination in the United Kingdom was developed. This improves on previous models by 1) allowing for 2 different mechanisms of action for Rotarix and RotaTeq, 2) using clinical trial data to understand these mechanisms, and 3) accounting for uncertainty by using Markov Chain Monte Carlo. RESULTS In the long run, Rotarix and RotaTeq are predicted to reduce the overall rotavirus incidence by 50% (39%-63%) and 44% (30%-62%), respectively but with an increase in incidence in primary school children and adults up to 25 y of age. The vaccines are estimated to give more protection than 1 or 2 natural infections. The duration of protection is highly uncertain but has only impact on the predicted reduction in rotavirus burden for values lower than 10 y. The 2 vaccine mechanism structures fit equally well with the clinical trial data. Long-term postvaccination dynamics cannot be predicted reliably with the data available. CONCLUSION Accounting for the joint uncertainty of several vaccine characteristics resulted in more insight into which of these are crucial for determining the impact of rotavirus vaccination. Data for up to at least 10 y postvaccination and covering older children and adults are crucial to address remaining questions on the impact of widespread rotavirus vaccination.
Collapse
Affiliation(s)
- Joke Bilcke
- Centre for Health Economics Research & Modeling of Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium (JB, LW),Department of Infectious Diseases Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK (JB, HJ, WJE, MJ)
| | - Ruth Chapman
- Modelling and Economics Unit, Public Health England, London, UK (RC, MJ)
| | - Christina Atchison
- Department of Primary Care and Public Health, School of Public Health, Imperial College, London, UK (CA)
| | - Deborah Cromer
- Complex Systems in Biology Group, Centre for Vascular Research, UNSW Australia, Sydney, New South Wales, Australia (DC)
| | - Helen Johnson
- Department of Infectious Diseases Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK (JB, HJ, WJE, MJ)
| | - Lander Willem
- Centre for Health Economics Research & Modeling of Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium (JB, LW),Department of Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium (LW),Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Diepenbeek, Belgium (LW)
| | - Martin Cox
- Department of Applied Sciences and Health, Coventry University, Coventry, UK (MC)
| | - William John Edmunds
- Department of Infectious Diseases Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK (JB, HJ, WJE, MJ)
| | - Mark Jit
- Department of Infectious Diseases Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK (JB, HJ, WJE, MJ),Modelling and Economics Unit, Public Health England, London, UK (RC, MJ)
| |
Collapse
|
223
|
Lee YY, Lee EB, Choi KH. Difference in the distribution of onset age of intussusception after rotavirus vaccination and according to the type of rotavirus vaccine: single medical center study. Yeungnam Univ J Med 2015. [DOI: 10.12701/yujm.2015.32.2.80] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Yun Young Lee
- Department of Pediatrics, College of Medicine, Yeungnam University, Daegu, Korea
| | - Eung Bin Lee
- Department of Pediatrics, College of Medicine, Yeungnam University, Daegu, Korea
| | - Kwang Hae Choi
- Department of Pediatrics, College of Medicine, Yeungnam University, Daegu, Korea
| |
Collapse
|
224
|
Correlates of protection against human rotavirus disease and the factors influencing protection in low-income settings. Mucosal Immunol 2015; 8:1-17. [PMID: 25465100 DOI: 10.1038/mi.2014.114] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 10/16/2014] [Indexed: 02/04/2023]
Abstract
Rotaviruses (RV) are the leading cause of gastroenteritis in infants and children worldwide and are associated with high mortality predominately in low-income settings. The virus is classified into G and P serotypes and further into P genotypes based on differences in the surface-exposed proteins VP7 and VP4, respectively. Infection results in a variable level of protection from subsequent reinfection and disease. This protection is predominantly homotypic in some settings, whereas broader heterotypic protection is reported in other cohorts. Two antigenically distinct oral RV vaccines are licensed and are being rolled out widely, including in resource-poor setting, with funding provided by the GAVI alliance. First is a monovalent vaccine derived from a live-attenuated human RV strain, whereas the second is a pentavalent bovine-human reassortment vaccine. Both vaccines are highly efficacious in high-income settings, but greatly reduced levels of protection are reported in low-income countries. Here, the current challenges facing mucosal immunologists and vaccinologists aiming to define immunological correlates and to understand the variable levels of protection conferred by these vaccines in humans is considered. Such understanding is critical to maximize the public health impact of the current vaccines and also to the development of the next generation of RV vaccines, which are needed.
Collapse
|
225
|
Ramani S, Atmar RL. Acute Gastroenteritis Viruses. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00057-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
226
|
Dormitzer PR. Rotaviruses. MANDELL, DOUGLAS, AND BENNETT'S PRINCIPLES AND PRACTICE OF INFECTIOUS DISEASES 2015:1854-1864.e4. [DOI: 10.1016/b978-1-4557-4801-3.00152-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
227
|
Strain diversity plays no major role in the varying efficacy of rotavirus vaccines: An overview. INFECTION GENETICS AND EVOLUTION 2014; 28:561-71. [DOI: 10.1016/j.meegid.2014.10.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 09/22/2014] [Accepted: 10/09/2014] [Indexed: 12/22/2022]
|
228
|
Cunliffe N, Zaman K, Rodrigo C, Debrus S, Benninghoff B, Pemmaraju Venkata S, Han HH. Early exposure of infants to natural rotavirus infection: a review of studies with human rotavirus vaccine RIX4414. BMC Pediatr 2014; 14:295. [PMID: 25433534 PMCID: PMC4261882 DOI: 10.1186/s12887-014-0295-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 11/11/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Rotaviruses are the leading cause of severe acute gastroenteritis in children aged <5 years worldwide. A live attenuated human rotavirus vaccine, RIX4414 has been developed to reduce the global disease burden associated with rotavirus gastroenteritis. Serum anti-rotavirus immunoglobulin A (IgA) antibody measured in unvaccinated infants during clinical trials of RIX4414 reflects natural rotavirus exposure, and may inform the optimal timing for rotavirus vaccination. METHODS We reviewed phase II and III randomized, placebo-controlled clinical trials conducted by GlaxoSmithKline Vaccines, Wavre, Belgium between 2000 and 2008 which used the commercial formulation of RIX4414 lyophilized vaccine. We included trials for which demographic data and pre-dose-1 and post-last-dose anti-rotavirus IgA antibody status were available from placebo recipients. RESULTS Sixteen clinical trials met the inclusion criteria. The studies were conducted across Africa (N = 3), Asia (N = 4), Latin America (N = 4), Europe (N = 4) and North America (N = 1). Overall, 46,398 infants were enrolled and among these, 20,099 received placebo. The mean age at pre-dose-1 time point ranged from 6.4 - 12.2 weeks while the mean age at post-last-dose time point ranged from 13.5 - 19.6 weeks. The anti-RV IgA seropositivity rates at both time points were higher in less developed countries of Africa, Asia and Latin America (pre-dose-1: 2.1%-26.3%; post-last-dose: 6.3%-34.8%) when compared to more developed countries of Asia, Europe and North America (pre-dose-1: 0%-9.4%; post-last-dose: 0%-21.3%), indicating that rotavirus infections occurred at a younger age in these regions. CONCLUSION Exposure to rotavirus infection occurred early in life among infants in most geographical settings, especially in developing countries. These data emphasize the importance of timely rotavirus vaccination within the Expanded Program on Immunization schedule to maximize protection.
Collapse
Affiliation(s)
| | | | - Carlos Rodrigo
- />Germans Trias i Pujol University Hospital, Universidad Autónoma de barcelona, Barcelona, Spain
| | | | | | | | - Htay-Htay Han
- />GlaxoSmithKline Vaccines, 2301 Renaissance Boulevard, King of Prussia, PA 19406 U.S.A
| |
Collapse
|
229
|
Mladenova Z, Nawaz S, Ganesh B, Iturriza-Gomara M. Increased detection of G3P[9] and G6P[9] rotavirus strains in hospitalized children with acute diarrhea in Bulgaria. INFECTION GENETICS AND EVOLUTION 2014; 29:118-26. [PMID: 25461849 DOI: 10.1016/j.meegid.2014.11.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/23/2014] [Accepted: 11/12/2014] [Indexed: 11/30/2022]
Abstract
Rotavirus severe disease in children is now vaccine-preventable and the roll-out of vaccination programs globally is expected to make a significant impact in the reduction of morbidity and mortality in children <5 years of age. Rotavirus is also a pathogen of other mammals and birds, and its segmented RNA genome can lead to the emergence of new or unusual strains in human population via interspecies transmission and reassortment events. Despite the efficacy and impact of rotavirus vaccine in preventing severe diarrhea, the correlates of protection remain largely unknown. Therefore, rotavirus strain surveillance before, during and after the introduction of immunization programs remains a crucial for monitoring rotavirus vaccine efficacy and impact. In this context, molecular characterization of 1323 Bulgarian rotavirus strains collected between June 2010 and May 2013 was performed. A total of 17 strains of interest were analyzed by partial sequence analysis. Twelve strains were characterized as G3P[9] and G6P[9] of potential animal origin. Phylogenetic analysis and comparisons with the same specificity strains detected sporadically between 2006 and 2010 revealed the constant circulation of these unusual human strains in Bulgaria, although in low prevalence, and their increased potential for person-to-person spread.
Collapse
Affiliation(s)
- Zornitsa Mladenova
- National Center for Infectious and Parasitic Diseases, Sofia, Bulgaria; Public Health of England, London, United Kingdom.
| | | | | | - Miren Iturriza-Gomara
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
230
|
Groome MJ, Page N, Cortese MM, Moyes J, Zar HJ, Kapongo CN, Mulligan C, Diedericks R, Cohen C, Fleming JA, Seheri M, Mphahlele J, Walaza S, Kahn K, Chhagan M, Steele AD, Parashar UD, Zell ER, Madhi SA. Effectiveness of monovalent human rotavirus vaccine against admission to hospital for acute rotavirus diarrhoea in South African children: a case-control study. THE LANCET. INFECTIOUS DISEASES 2014; 14:1096-1104. [DOI: 10.1016/s1473-3099(14)70940-5] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
231
|
Abstract
Vaccines are now available to combat rotavirus, the most common cause of severe diarrhea among children worldwide. We review clinical trial data for available rotavirus vaccines and summarize postlicensure data on effectiveness, impact, and safety from countries routinely using these vaccines in national programs. In these countries, rotavirus vaccines have reduced all-cause diarrhea and rotavirus hospitalizations by 17%-55% and 49%-92%, respectively, and all-cause diarrhea deaths by 22%-50% in some settings. Indirect protection of children who are age-ineligible for rotavirus vaccine has also been observed in some high and upper middle income countries. Experience with routine use of rotavirus vaccines in lower middle income countries has been limited to date, but vaccine introductions in such countries have been increasing in recent years. The risk-benefit analysis of rotavirus vaccines is extremely favorable but other strategies to improve the effectiveness of the vaccine, particularly in lower middle income settings, should be considered.
Collapse
Affiliation(s)
- Jacqueline E Tate
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Umesh D Parashar
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
232
|
Matthijnssens J, Zeller M, Heylen E, De Coster S, Vercauteren J, Braeckman T, Van Herck K, Meyer N, PirÇon JY, Soriano-Gabarro M, Azou M, Capiau H, De Koster J, Maernoudt AS, Raes M, Verdonck L, Verghote M, Vergison A, Van Damme P, Van Ranst M. Higher proportion of G2P[4] rotaviruses in vaccinated hospitalized cases compared with unvaccinated hospitalized cases, despite high vaccine effectiveness against heterotypic G2P[4] rotaviruses. Clin Microbiol Infect 2014; 20:O702-10. [DOI: 10.1111/1469-0691.12612] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/16/2014] [Accepted: 02/25/2014] [Indexed: 12/26/2022]
|
233
|
Zelman M, Sanford C, Neatby A, Halperin BA, MacDougall D, Rowswell C, Langley JM, Halperin SA. Implementation of a universal rotavirus vaccination program: comparison of two delivery systems. BMC Public Health 2014; 14:908. [PMID: 25182067 PMCID: PMC4165993 DOI: 10.1186/1471-2458-14-908] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 08/26/2014] [Indexed: 11/17/2022] Open
Abstract
Background Rotavirus vaccine is recommended for all infants in Canada. To evaluate the logistics of implementing a universal rotavirus vaccination program, we compared the effectiveness of program implementation in jurisdictions with either a physician-administered or public health nurse-administered program. Methods All infants born between October 1, 2010 and September 30, 2012 in Prince Edward Island and Nova Scotia’s Capital District Health Authority were eligible for the vaccination program. A universal rotavirus vaccination program was implemented and delivered in public health clinics in Prince Edward Island and in physicians’ offices in Nova Scotia. Results Engagement of vaccinators in delivery of the universal vaccination program was more successful in Prince Edward Island than in Nova Scotia. Vaccine coverage rates rose rapidly in Prince Edward Island, exceeding 90% for both doses within 3 months and remaining at those levels over the two-year program. In contrast, coverage rates in Nova Scotia rose more slowly and never exceeded 40% during the two years. Access to coverage data was more timely and accurate in Prince Edward Island than Nova Scotia. Conclusion A universal rotavirus vaccination program delivered through public health clinics achieved more rapid and higher levels of coverage than a program administered through physicians’ offices. Trial registration NCT01273077.
Collapse
|
234
|
Weidemann F, Dehnert M, Koch J, Wichmann O, Höhle M. Modelling the epidemiological impact of rotavirus vaccination in Germany – A Bayesian approach. Vaccine 2014; 32:5250-7. [DOI: 10.1016/j.vaccine.2014.06.090] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 05/23/2014] [Accepted: 06/11/2014] [Indexed: 01/17/2023]
|
235
|
Desselberger U. Rotaviruses. Virus Res 2014; 190:75-96. [PMID: 25016036 DOI: 10.1016/j.virusres.2014.06.016] [Citation(s) in RCA: 284] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 06/26/2014] [Accepted: 06/26/2014] [Indexed: 01/12/2023]
|
236
|
Characterization of G2P[4] rotavirus strains causing outbreaks of gastroenteritis in the Northern Territory, Australia, in 1999, 2004 and 2009. INFECTION GENETICS AND EVOLUTION 2014; 28:434-45. [PMID: 25152486 DOI: 10.1016/j.meegid.2014.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 08/01/2014] [Accepted: 08/11/2014] [Indexed: 01/18/2023]
Abstract
Outbreaks of rotavirus diarrhea cause a large disease burden in the Alice Springs region of the Northern Territory, Australia. The introduction of the rotavirus vaccine Rotarix® has been associated with an increase in detection of G2P[4] strains in many countries. However, G2P[4] emergence has also been observed in vaccine-naive countries, suggesting a general global increase in the circulation of G2P[4] strains. A G2P[4] rotavirus outbreak occurred in 2009, 28 months after the introduction of the Rotarix® vaccine and 43 children were hospitalized. Pre-vaccine introduction, G2P[4] strains were observed associated with large outbreaks in 1999 and 2004. To determine the genetic relationship between these strains whole genome sequence analysis was conducted on representative strains from each of the G2P[4] outbreaks, in 1999, 2004 and 2009. Phylogenetic analysis revealed the majority of genes from 2009 outbreak strain clustered with contemporary global strains, while the VP7 gene clustered with contemporary and older strains and was antigenically distinct to the majority of contemporary global G2P[4] strains; suggesting the strain was an intragenogroup reassortant. The 1999 and 2009 strains appear to share similar evolutionary origins, and both had a high degree of genetic identity to previously identified Australian and global strains. Conversely, the 2004 outbreak strain was more divergent in comparison to Australian and global strains. The 1999 and 2004 outbreaks likely occurred due to the accumulation of immunologically naïve children in the population following low levels of G2P[4] rotavirus disease in the community in the years prior to each outbreak. The 2009 outbreak was associated with moderate vaccine coverage in the population and vaccine efficacy against the strain was low. The circulation of this unusual strain in the population combined with low vaccine coverage and diminished vaccine efficacy likely contributed to the outbreak occurring in this population.
Collapse
|
237
|
Rao TS, Arora R, Khera A, Tate JE, Parashar U, Kang G. Insights from global data for use of rotavirus vaccines in India. Vaccine 2014; 32 Suppl 1:A171-8. [PMID: 25091672 PMCID: PMC12011198 DOI: 10.1016/j.vaccine.2014.03.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Rotavirus vaccines are being introduced in several low- and middle-income countries across the world with and without support from the GAVI Alliance. India has the highest disease burden of rotavirus based on morbidity and mortality estimates and several indigenous vaccine manufacturers are developing rotavirus vaccines. One candidate has undergone phase III testing and others have completed evaluation in phase II. Global data on licensed vaccine performance in terms of impact on disease, strain diversity, safety and cost-effectiveness has been reviewed to provide a framework for decision making in India.
Collapse
Affiliation(s)
- T S Rao
- Department of Biotechnology, Ministry of Science and Technology, Government of India, CGO Complex, New Delhi, India
| | - Rashmi Arora
- Division of Epidemiology and Communicable Diseases, Indian Council for Medical Research, Ansari Nagar, New Delhi, India
| | - Ajay Khera
- Child Health and Immunization, Ministry of Health and Family Welfare, Nirman Bhawan, New Delhi, India
| | - Jacqueline E Tate
- Viral Gastroenteritis Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Umesh Parashar
- Viral Gastroenteritis Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Gagandeep Kang
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore 632004, Tamil Nadu, India.
| |
Collapse
|
238
|
Dhingra M, Kundu R, Gupta M, Kanungo S, Ganguly N, Singh M, Bhattacharya M, Ghosh R, Kumar R, Sur D, Chadha S, Saluja T. Evaluation of safety and immunogenicity of a live attenuated tetravalent (G1–G4) Bovine-Human Reassortant Rotavirus vaccine (BRV-TV) in healthy Indian adults and infants. Vaccine 2014; 32 Suppl 1:A117-23. [DOI: 10.1016/j.vaccine.2014.03.069] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
239
|
Effect of withholding breastfeeding on the immune response to a live oral rotavirus vaccine in North Indian infants. Vaccine 2014; 32 Suppl 1:A134-9. [DOI: 10.1016/j.vaccine.2014.04.078] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
240
|
Leshem E, Lopman B, Glass R, Gentsch J, Bányai K, Parashar U, Patel M. Distribution of rotavirus strains and strain-specific effectiveness of the rotavirus vaccine after its introduction: a systematic review and meta-analysis. THE LANCET. INFECTIOUS DISEASES 2014; 14:847-56. [PMID: 25082561 DOI: 10.1016/s1473-3099(14)70832-1] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Concerns exist about whether monovalent (RV1) and pentavalent (RV5) rotavirus vaccines provide adequate protection against diverse strains and whether vaccine introduction will lead to selective pressure. We aimed to investigate the distribution of rotavirus strains and strain-specific rotavirus vaccine effectiveness after vaccine introduction. METHODS We did a systematic review of published work to assess the strain-specific effectiveness of RV1 and RV5 rotavirus vaccines. We classified strains as homotypic, partly heterotypic, and fully heterotypic based on the amount of antigen-matching between strain and vaccine. When studies reported vaccine effectiveness against single antigens (G-type or P-type), we categorised them as either single-antigen vaccine type or single-antigen non-vaccine type. Our primary outcome was strain-specific vaccine effectiveness, comparing effectiveness of homotypic strains with fully or partly heterotypic strains. A secondary outcome was the prevalence of rotavirus strains after vaccine introduction. We estimated pooled odds ratios using random-effect regression models, stratified by country income level and vaccine type, and tested for differences in strain-specific vaccine effectiveness. We assessed strain distribution trends from surveillance reports. FINDINGS In high-income countries, RV1 pooled vaccine effectiveness was 94% (95% CI 80-98) against homotypic strains, 71% (39-86) against partly heterotypic strains, and 87% (76-93) against fully heterotypic strains. In middle-income settings, respective pooled data were 59% (36-73), 72% (58-81), and 47% (28-61). In high-income countries, RV5 vaccine effectiveness was 83% (78-87) against homotypic strains, 82% (70-89) against single-antigen vaccine type strains, 82% (70-89) against partly heterotypic strains, and 75% (47-88) against single-antigen non-vaccine type strains. In middle-income settings, RV5 vaccine effectiveness was 70% (58-78) against single-antigen vaccine type strains, 37% (10-56) against partly heterotypic strains, and 87% (38-97) against single-antigen non-vaccine type strains. No difference was noted in vaccine effectiveness for either RV1 or RV5 in any setting (all p>0·05). Prevalent strains in countries using RV1 were G2P[4] (2198 of 4428, 50%) and G1P[8] (953, 22%), and those in countries using RV5 were G1P[8] (1280 of 3875, 33%) and G2P[4] (1169, 30%). Sustained predominance of a single strain was not recorded. INTERPRETATION RV1 and RV5 exert similar effectiveness against homotypic and heterotypic rotavirus strains. Persistence of specific strains was not recorded, suggesting vaccine-induced selective pressure did not occur. Expansion of rotavirus surveillance efforts to low-income countries and ongoing surveillance are crucial to identify emergence of new strains and to assess strain-specific vaccine effectiveness in various settings. FUNDING None.
Collapse
Affiliation(s)
- Eyal Leshem
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA; Epidemic Intelligence Service, Division of Scientific Education and Professional Development, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Ben Lopman
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Roger Glass
- Fogarty International Center, National Institutes of Health, Bethesda, MD, USA
| | - Jon Gentsch
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Krisztián Bányai
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Umesh Parashar
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Manish Patel
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
241
|
Rha B, Tate JE, Weintraub E, Haber P, Yen C, Patel M, Cortese MM, DeStefano F, Parashar UD. Intussusception following rotavirus vaccination: an updated review of the available evidence. Expert Rev Vaccines 2014; 13:1339-48. [DOI: 10.1586/14760584.2014.942223] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
242
|
Donato CM, Cowley D, Snelling TL, Akopov A, Kirkness EF, Kirkwood CD. Characterization of a G1P[8] rotavirus causing an outbreak of gastroenteritis in the Northern Territory, Australia, in the vaccine era. Emerg Microbes Infect 2014; 3:e47. [PMID: 26038746 PMCID: PMC4126178 DOI: 10.1038/emi.2014.47] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/01/2014] [Accepted: 05/05/2014] [Indexed: 02/01/2023]
Abstract
In 2010, a large outbreak of rotavirus gastroenteritis occurred in the Alice Springs region of the Northern Territory, Australia. The outbreak occurred 43 months after the introduction of the G1P[8] rotavirus vaccine Rotarix®. Forty-three infants were hospitalized during the outbreak and analysis of fecal samples from each infant revealed a G1P[8] rotavirus strain. The outbreak strain was adapted to cell culture and neutralization assays were performed using VP7 and VP4 neutralizing monoclonal antibodies. The outbreak strain exhibited a distinct neutralization resistance pattern compared to the Rotarix® vaccine strain. Whole genome sequencing of the 2010 outbreak virus strain demonstrated numerous amino acid differences compared to the Rotarix® vaccine strain in the characterized neutralization epitopes of the VP7 and VP4 proteins. Phylogenetic analysis of the outbreak strain revealed a close genetic relationship to global strains, in particular RVA/Human-wt/BEL/BE0098/2009/G1P[8] and RVA/Human-wt/BEL/BE00038/2008/G1P[8] for numerous genes. The 2010 outbreak strain was likely introduced from a globally circulating population of strains rather than evolving from an endemic Australian strain. The outbreak strain possessed antigenic differences in the VP7 and VP4 proteins compared to the Rotarix® vaccine strain. The outbreak was associated with moderate vaccine coverage and possibly low vaccine take in the population.
Collapse
Affiliation(s)
- Celeste M Donato
- Murdoch Childrens Research Institute, Royal Children's Hospital , Melbourne 3052, Victoria, Australia ; La Trobe University , Bundoora 3086, Victoria, Australia
| | - Daniel Cowley
- Murdoch Childrens Research Institute, Royal Children's Hospital , Melbourne 3052, Victoria, Australia
| | - Thomas L Snelling
- Menzies School of Health Research and Charles Darwin University , Casuarina 0810, Northern Territory, Australia
| | - Asmik Akopov
- The J Craig Venter Institute , Rockville, MD 20850, USA
| | | | - Carl D Kirkwood
- Murdoch Childrens Research Institute, Royal Children's Hospital , Melbourne 3052, Victoria, Australia ; La Trobe University , Bundoora 3086, Victoria, Australia
| |
Collapse
|
243
|
Huppertz HI, Borte M, Schuster V, Giaquinto C, Vesikari T. Report of the Third European Expert Meeting on Rotavirus Vaccination: Progress in rotavirus universal mass vaccination in Europe. Vaccine 2014; 32:4243-8. [DOI: 10.1016/j.vaccine.2014.05.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 05/09/2014] [Indexed: 11/26/2022]
|
244
|
Bhandari N, Rongsen-Chandola T, Bavdekar A, John J, Antony K, Taneja S, Goyal N, Kawade A, Kang G, Rathore SS, Juvekar S, Muliyil J, Arya A, Shaikh H, Abraham V, Vrati S, Proschan M, Kohberger R, Thiry G, Glass R, Greenberg HB, Curlin G, Mohan K, Harshavardhan GVJA, Prasad S, Rao TS, Boslego J, Bhan MK. Efficacy of a monovalent human-bovine (116E) rotavirus vaccine in Indian infants: a randomised, double-blind, placebo-controlled trial. Lancet 2014; 383:2136-43. [PMID: 24629994 PMCID: PMC4532697 DOI: 10.1016/s0140-6736(13)62630-6] [Citation(s) in RCA: 217] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Rotavirus is the most common cause of severe dehydrating gastroenteritis in developing countries. Safe, effective, and affordable rotavirus vaccines are needed in these countries. We aimed to assess the efficacy and tolerability of a monovalent human-bovine rotavirus vaccine for severe rotavirus gastroenteritis in low-resource urban and rural settings in India. METHODS We did a randomised double-blind, placebo-controlled, multicentre trial at three sites in Delhi (urban), Pune (rural), and Vellore (urban and rural) between March 11, 2011, and Nov 5, 2012. Infants aged 6-7 weeks were randomly assigned (2:1), via a central interactive voice or web response system with a block size of 12, to receive either three doses of oral human-bovine natural reassortant vaccine (116E) or placebo at ages 6-7 weeks, 10 weeks, and 14 weeks. Infants' families, study investigators, paediatricians in referral hospitals, laboratory staff, and committee members were all masked to treatment allocation. The primary outcome was incidence of severe rotavirus gastroenteritis (≥11 on the Vesikari scale). Efficacy outcomes and adverse events were ascertained through active surveillance. Analysis was by intention to treat and per protocol. The trial is registered with Clinical Trial Registry-India (CTRI/2010/091/000102) and ClinicalTrials.gov (NCT01305109). FINDINGS 4532 infants were assigned to receive the 116E vaccine and 2267 to receive placebo, of whom 4354 (96%) and 2187 (96%) infants, respectively, were included in the primary per-protocol efficacy analysis. 71 events of severe rotavirus gastroenteritis were reported in 4752 person-years in infants in the vaccine group compared with 76 events in 2360 person-years in those in the placebo group; vaccine efficacy against severe rotavirus gastroenteritis was 53·6% (95% CI 35·0-66·9; p=0·0013) and 56·4% (36·6-70·1; p<0·0001) in the first year of life. The number of infants needed to be immunised to prevent one severe rotavirus gastroenteritis episode was 55 (95% CI 37-97). The incidence of severe rotavirus gastroenteritis per 100 person-years was 1·5 in the vaccine group and 3·2 in the placebo group, with an incidence rate ratio of 0·46 (95% CI 0·33-0·65). Prevalence of immediate, solicited, and serious adverse events was similar in both groups. One case of urticaria in the vaccine group and one each of acute gastroenteritis and suspected sepsis in the placebo group were regarded as related to the study product. We recorded six cases of intussusception in the vaccine group and two in the placebo group, all of which happened after the third dose. 25 (<1%) infants in the vaccine group and 17 (<1%) in the placebo group died; no death was regarded as related to the study product. INTERPRETATION Monovalent human-bovine (116E) rotavirus vaccine is effective and well tolerated in Indian infants. FUNDING Department of Biotechnology and the Biotechnology Industry Research Assistance Council, Government of India; Bill & Melinda Gates Foundation to PATH, USA; Research Council of Norway; UK Department for International Development; National Institutes of Health, Bethesda, USA; and Bharat Biotech International, Hyderabad, India.
Collapse
Affiliation(s)
- Nita Bhandari
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | | | | | - Jacob John
- Christian Medical College, Vellore, Tamil Nadu, India
| | | | - Sunita Taneja
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | - Nidhi Goyal
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | - Anand Kawade
- KEM Hospital Research Centre, Pune, Maharashtra, India
| | | | - Sudeep Singh Rathore
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | | | | | - Alok Arya
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | - Hanif Shaikh
- KEM Hospital Research Centre, Pune, Maharashtra, India
| | - Vinod Abraham
- Christian Medical College, Vellore, Tamil Nadu, India
| | - Sudhanshu Vrati
- Translational Health Science and Technology Institute, Gurgaon, Haryana, India
| | | | | | - Georges Thiry
- Advancing Rotavirus Vaccines Development Project, PATH, France
| | - Roger Glass
- National Institutes of Health, Bethesda, MD, USA
| | | | | | - Krishna Mohan
- Bharat Biotech International, Genome Valley, Andhra Pradesh, India
| | | | - Sai Prasad
- Bharat Biotech International, Genome Valley, Andhra Pradesh, India
| | - T S Rao
- Department of Biotechnology, Government of India, India
| | | | | |
Collapse
|
245
|
Anderson EJ. Time to Begin a New Chapter and Expand Rotavirus Immunization. Clin Infect Dis 2014; 59:982-6. [DOI: 10.1093/cid/ciu475] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
246
|
Abstract
Rotavirus gastroenteritis is a vaccine-preventable disease that confers a high medical and economic burden in more developed countries and can be fatal in less developed countries. Two vaccines with high efficacy and good safety profiles were approved and made available in Europe in 2006. We present an overview of the status of rotavirus vaccination in Europe. We discuss the drivers (including high effectiveness and effect of universal rotavirus vaccination) and barriers (including low awareness of disease burden, perception of unfavourable cost-effectiveness, and potential safety concerns) to the implementation of universal rotavirus vaccination in Europe. By February, 2014, national universal rotavirus vaccination had been implemented in Belgium, Luxembourg, Austria, Finland, Greece, Luxembourg, Norway, and the UK. Four other German states have issued recommendations and reimbursement is provided by sickness funds. Other countries were at various stages of recommending or implementing universal rotavirus vaccination.
Collapse
|
247
|
Azegami T, Yuki Y, Kiyono H. Challenges in mucosal vaccines for the control of infectious diseases. Int Immunol 2014; 26:517-28. [DOI: 10.1093/intimm/dxu063] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
248
|
Anca IA, Furtunescu FL, Pleşca D, Streinu-Cercel A, Rugină S, Holl K. Hospital-based surveillance to estimate the burden of rotavirus gastroenteritis in children below five years of age in Romania. Germs 2014; 4:30-40. [PMID: 24967217 DOI: 10.11599/germs.2014.1053] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 04/19/2014] [Accepted: 04/28/2014] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Rotavirus (RV) is a leading cause of acute gastroenteritis (AGE), affecting 95% of children below five years of age. METHODS In this prospective, multi-center study, children below five years of age who were hospitalized or those who visited the emergency room (ER) due to AGE or who developed AGE at least 48 hours after hospitalization (nosocomial infection) and had a RV-positive stool sample were included (n=1,222). RV-positive samples were genotyped by reverse-transcriptase polymerase chain reaction. RESULTS RV test results were available for 1,212 children (hospitalizations [n=677], ER visits [n=398] and nosocomial AGE cases [n=137]). Proportions of rotavirus gastroenteritis (RVGE) hospitalizations and ER visits were 51.70% (350/677; 95%CI: 47.86-55.52) and 36.18% (144/398; 95%CI: 31.45-41.12), respectively. Overall, 45.95% (494/1075) of all community-acquired AGE cases were due to RV. High numbers of RVGE cases were recorded between January and March. Most common genotypes were G9P[8] (34.27%) followed by G4P[8] (25.83%) and G1P[8] (23.02%). Of all community-acquired RVGE cases, the highest number of cases was observed in children aged 12-23 months. Median duration of hospitalization among RV-positive subjects was six days (range: 2-31 days). Incidence of nosocomial RVGE was 0.52 (95%CI: 0.45-0.60) cases per 1,000 child-days hospitalization. Median duration for additional hospitalization due to nosocomial RVGE was five days (range: 1-10). The highest burden of nosocomial RVGE was observed in children aged 12-23 months (42.34%, 58/137). Our findings confirm a high burden of acute RVGE disease in Romania and provide useful data to support the implementation of RV vaccination in Romania. TRIAL REGISTRATION NCT01253967.
Collapse
Affiliation(s)
- Ioana Alina Anca
- MD, PhD, Carol Davila University of Medicine and Pharmacy; Institute For Mother and Child Care, Bucharest 020395, Romania
| | - Florentina Ligia Furtunescu
- MD, PhD, Department of Complementary Sciences, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest 050463, Romania
| | - Doina Pleşca
- MD, PhD, Carol Davila University of Medicine and Pharmacy; Children's Clinical Hospital "Dr. Victor Gomoiu", Bucharest 022102, Romania
| | - Adrian Streinu-Cercel
- MD, PhD, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; National Institute for Infectious Diseases "Prof. Dr. Matei Balş", Bucharest 021105, Romania
| | - Sorin Rugină
- MD, PhD, Ovidius University, Faculty of Medicine, Infectious Diseases Clinic, Constanţa 900709, Romania
| | | |
Collapse
|
249
|
Clark A, Jit M, Andrews N, Atchison C, Edmunds WJ, Sanderson C. Evaluating the potential risks and benefits of infant rotavirus vaccination in England. Vaccine 2014; 32:3604-10. [DOI: 10.1016/j.vaccine.2014.04.082] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/21/2014] [Accepted: 04/23/2014] [Indexed: 12/16/2022]
|
250
|
Magagula NB, Esona MD, Nyaga MM, Stucker KM, Halpin RA, Stockwell TB, Seheri ML, Steele AD, Wentworth DE, Mphahlele MJ. Whole genome analyses of G1P[8] rotavirus strains from vaccinated and non-vaccinated South African children presenting with diarrhea. J Med Virol 2014; 87:79-101. [PMID: 24841697 DOI: 10.1002/jmv.23971] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2014] [Indexed: 12/18/2022]
Abstract
Group A rotaviruses (RVAs) are the leading cause of severe gastroenteritis and eventually death among infants and young children worldwide, and disease prevention and management through vaccination is a public health priority. In August 2009, Rotarix™ was introduced in the South African Expanded Programme on Immunisation. As a result, substantial reductions in RVA disease burden have been reported among children younger than 5 years old. Rotavirus strain surveillance post-vaccination is crucial to, inter alia, monitor and study the evolution of vaccine escape strains. Here, full-genome sequence data for the 11 gene segments from 11 South African G1P[8] rotavirus strains were generated, including 5 strains collected from non-vaccinated children during the 2004-2009 rotavirus seasons and 6 strains collected from vaccinated children during the 2010 rotavirus season. These data were analyzed to gain insights into the overall genetic makeup and evolution of South African G1P[8] rotavirus strains and to compare their genetic backbones with those of common human Wa-like RVAs from other countries, as well as with the Rotarix™ and RotaTeq™ G1P[8] vaccine components. All 11 South African G1P[8] strains revealed a complete Wa-like genotype constellation of G1-P[8]-I1-R1-C1-M1-A1-N1-T1-E1-H1. On the basis of sequence similarities, the South African G1P[8] strains (with the exception of strain RVA/Human-wt/ZAF/1262/2004/G1P[8]) were closely related to each other (96-100% identity in all gene segments). Comparison to the Rotarix™ and RotaTeq™ G1P[8] vaccine components revealed a moderate nucleotide identity of 89-96% and 93-95%, respectively. The results indicated that none of the gene segments of these 11 South African G1P[8] strains were vaccine-derived. This study illustrates that large-scale next generation sequencing will provide crucial information on the influence of the vaccination program on evolution of rotavirus strains. This is the first report to describe full genomic analyses of G1P[8] RVA strains collected from both non-vaccinated and vaccinated children in South Africa.
Collapse
Affiliation(s)
- Nonkululeko B Magagula
- Medical Research Council/Diarrhoeal Pathogens Research Unit, Department of Virology, University of Limpopo, Medunsa Campus/National Health Laboratory Service, Pretoria, South Africa
| | | | | | | | | | | | | | | | | | | |
Collapse
|