301
|
Seheri LM, Page NA, Mawela MPB, Mphahlele MJ, Steele AD. Rotavirus vaccination within the South African Expanded Programme on Immunisation. Vaccine 2013; 30 Suppl 3:C14-20. [PMID: 22939015 DOI: 10.1016/j.vaccine.2012.04.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 04/03/2012] [Accepted: 04/04/2012] [Indexed: 01/30/2023]
Abstract
Diarrhoeal diseases are ranked the third major cause of childhood mortality in South African children less than 5 years, where the majority of deaths are among black children. Acute severe dehydrating rotavirus diarrhoea remains an important contributor towards childhood mortality and morbidity and has been well documented in South Africa. As the preventive strategy to control rotavirus diarrhoea, South Africa became the first country in the WHO African Region to adopt the rotavirus vaccine in the national childhood immunisation programme in August 2009. The rotavirus vaccine in use, Rotarix, GSK Biologicals, is given at 6 and 14 weeks of age, along with other vaccines as part of Expanded Programme on Immunisation (EPI). Studies which facilitated the introduction of rotavirus vaccine in South Africa included the burden of rotavirus disease and strain surveillance, economic burden of rotavirus infection and clinical trials to assess the safety and efficacy of vaccine candidates. This paper reviews the epidemiology of rotavirus in South Africa, outlines some of the steps followed to introduce rotavirus vaccine in the EPI, and highlights the early positive impact of vaccination in reducing the rotavirus burden of disease based on the post-marketing surveillance studies at Dr George Mukhari hospital, a sentinel site at University of Limpopo teaching hospital in Pretoria, South Africa, which has conducted rotavirus surveillance for >20 years.
Collapse
Affiliation(s)
- L Mapaseka Seheri
- MRC/UL Diarrhoeal Pathogens Research Unit, Department of Virology, Medunsa Campus, University of Limpopo/National Health Laboratory Service, Pretoria, South Africa.
| | | | | | | | | |
Collapse
|
302
|
Standaert B, Gomez JA, Raes M, Debrus S, Velázquez FR, Postma MJ. Impact of rotavirus vaccination on hospitalisations in Belgium: comparing model predictions with observed data. PLoS One 2013; 8:e53864. [PMID: 23349754 PMCID: PMC3548809 DOI: 10.1371/journal.pone.0053864] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 12/04/2012] [Indexed: 11/23/2022] Open
Abstract
Background Published economic assessments of rotavirus vaccination typically use modelling, mainly static Markov cohort models with birth cohorts followed up to the age of 5 years. Rotavirus vaccination has now been available for several years in some countries, and data have been collected to evaluate the real-world impact of vaccination on rotavirus hospitalisations. This study compared the economic impact of vaccination between model estimates and observed data on disease-specific hospitalisation reductions in a country for which both modelled and observed datasets exist (Belgium). Methods A previously published Markov cohort model estimated the impact of rotavirus vaccination on the number of rotavirus hospitalisations in children aged <5 years in Belgium using vaccine efficacy data from clinical development trials. Data on the number of rotavirus-positive gastroenteritis hospitalisations in children aged <5 years between 1 June 2004 and 31 May 2006 (pre-vaccination study period) or 1 June 2007 to 31 May 2010 (post-vaccination study period) were analysed from nine hospitals in Belgium and compared with the modelled estimates. Results The model predicted a smaller decrease in hospitalisations over time, mainly explained by two factors. First, the observed data indicated indirect vaccine protection in children too old or too young for vaccination. This herd effect is difficult to capture in static Markov cohort models and therefore was not included in the model. Second, the model included a ‘waning’ effect, i.e. reduced vaccine effectiveness over time. The observed data suggested this waning effect did not occur during that period, and so the model systematically underestimated vaccine effectiveness during the first 4 years after vaccine implementation. Conclusions Model predictions underestimated the direct medical economic value of rotavirus vaccination during the first 4 years of vaccination by approximately 10% when assessing hospitalisation rates as compared with observed data in Belgium.
Collapse
|
303
|
Lu CY, Chang LY, Shao PL, Suryakiran PV, Han HH, Huang LM. Immunogenicity, reactogenicity, and safety of a human rotavirus vaccine, Rotarix, in Taiwanese infants who received a dose of hepatitis B immunoglobulin after birth. J Formos Med Assoc 2013; 112:574-7. [PMID: 24079716 DOI: 10.1016/j.jfma.2012.11.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 11/09/2012] [Accepted: 11/23/2012] [Indexed: 10/27/2022] Open
Abstract
UNLABELLED This Phase-IV study evaluated the human rotavirus (RV) vaccine Rotarix (RIX4414) to provide additional local clinical data to the Taiwan Food and Drug Association (NCT01198769). Healthy infants aged 6-12 weeks who were given a hepatitis B immunoglobulin (HBIg) dose after birth, received two doses of RIX4414 (0, 2-month schedule). Anti-RV IgA antibody concentrations were measured using ELISA. A total of 15 infants were enrolled, and included in the according-to-protocol cohort. The anti-RV IgA antibody seroconversion rate 2 months post-Dose 2 was 100% (95% confidence interval = 78.2-100) and the geometric mean concentration was 254.7 U/ml (95% confidence interval = 145.0-447.7). Two episodes of gastroenteritis were reported, and one stool sample was tested for RV, which was negative. No fatal serious adverse events were reported during the study period between November 2010 and April 2011. The two-dose regimen of RIX4414 was highly immunogenic and safe when administered to healthy Taiwanese infants who received a HBIg dose after birth. TRIAL REGISTRATION NUMBER NCT01198769.
Collapse
Affiliation(s)
- Chun-Yi Lu
- Department of Pediatrics, National Taiwan University Hospital, Taipei 100, Taiwan
| | | | | | | | | | | |
Collapse
|
304
|
Bernard S, Valiquette L, De Wals P, Nault V, Babakissa C, Cyr C, Boileau TC, Gagneur A. Burden of rotavirus disease: A population-based study in Eastern Townships, Quebec. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2013; 24:138-42. [PMID: 24421824 PMCID: PMC3852450 DOI: 10.1155/2013/919124] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Since July 2010, the National Advisory Committee on Immunization of Canada has recommended rotavirus vaccination for all healthy infants. However, before implementing this vaccine in routine health programs, Canadian provinces need to establish current epidemiological data on rotavirus-associated acute gastroenteritis (AGE). METHODS A retrospective cohort study of children <5 years of age with AGE from 2002 to 2008 was performed in Eastern Townships, Quebec (population in 2006: 298,780). Data were collected on visits to outpatient clinics, emergency department (ED) visits, hospitalizations (standard and short-stay units) and nosocomial AGE. The winter residual estimation and Brandt methods were used to estimate the proportion of AGE attributable to rotaviruses. RESULTS During the six-year study period, a total of 1435 hospitalizations, 3631 ED visits and 6220 ambulatory care visits were attributed to AGE. The specific rotavirus burden was estimated to be 449 to 666 for hospitalizations, 1050 to 1361 for ED visits and 1633 to 1687 for outpatient visits. The epidemic curve showed a periodicity with higher incidence in March and April. Short-stay unit hospitalizations represented 58% of all hospitalizations. The annual incidence rate of rotaviruses was estimated to be 50 to 74 per 10,000 children for hospitalizations, 117 to 152 per 10,000 children for ED visits and 182 to 188 per 10,000 children for outpatient visits. CONCLUSION Most available retrospective studies probably underestimate rotavirus-associated hospitalizations because they do not take into account short-stay unit hospitalizations. Furthermore, these data on emergency and outpatient visits provide an exhaustive appraisal of the rotavirus burden, which serves as crucial information for the evaluation of immunization programs.
Collapse
Affiliation(s)
- Sylvain Bernard
- Department of Pediatrics, University of Sherbrooke, Sherbrooke
- Center of clinical research Etienne le Bel, CHUS, Québec
| | - Louis Valiquette
- Center of clinical research Etienne le Bel, CHUS, Québec
- Department of Microbiology and Infectious Diseases, University of Sherbrooke, Sherbrooke
| | - Philippe De Wals
- Department of Social and Preventive Medicine, University of Laval, Québec City, Quebec
| | - Vincent Nault
- Department of Microbiology and Infectious Diseases, University of Sherbrooke, Sherbrooke
| | | | - Claude Cyr
- Department of Pediatrics, University of Sherbrooke, Sherbrooke
| | | | - Arnaud Gagneur
- Department of Pediatrics, University of Sherbrooke, Sherbrooke
- Center of clinical research Etienne le Bel, CHUS, Québec
| |
Collapse
|
305
|
Estimating and comparing the clinical and economic impact of paediatric rotavirus vaccination in Turkey using a simple versus an advanced model. Vaccine 2012; 31:979-86. [PMID: 23219433 DOI: 10.1016/j.vaccine.2012.11.071] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 11/20/2012] [Accepted: 11/25/2012] [Indexed: 11/20/2022]
Abstract
BACKGROUND The burden of rotavirus disease is high in Turkey, reflecting the large birth cohort (>1.2 million) and the risk of disease. Modelling can help to assess the potential economic impact of vaccination. We compared the output of an advanced model with a simple model requiring fewer data inputs. If the results are similar, this could be helpful for countries that have few data available. METHODS The advanced model was a previously published static Markov cohort model comparing costs and quality-adjusted life-year (QALY) outcomes of vaccination versus no vaccination. In contrast, the simple model used only a decision tree. Both models included data on demography, epidemiology, vaccine efficacy, resource use, unit costs, and utility scores from national databases and published papers. Only the perspective of the health care payer was considered in the analysis. The simple model had 23 variables, compared with 103 in the advanced model to allow additional comparisons of different vaccine types, dose schemes and vaccine waning. RESULTS With the same input data, both models showed that rotavirus vaccination in Turkey would improve health outcomes (fewer QALYs lost to rotavirus disease). The projected annual cost offsets were $29.9 million in the simple and $29.4 million in the advanced model. Sensitivity analysis indicated that in both models the main cost driver was disease incidence followed by cost for hospital care and medical visits. Vaccine efficacy had a smaller effect. CONCLUSION Both models reached similar conclusions. Both projected that rotavirus vaccination in Turkey would improve health outcomes and may result in savings in direct healthcare costs to offset the cost of vaccination. The analysis indicated that the simple model can produce meaningful economic results in conditions where few data are available.
Collapse
|
306
|
Rotavirus vaccination effectiveness: A case–case study in the EDICS project, Castellón (Spain). Vaccine 2012; 30:7536-40. [DOI: 10.1016/j.vaccine.2012.10.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 09/28/2012] [Accepted: 10/13/2012] [Indexed: 11/20/2022]
|
307
|
Fisman DN, Chan CH, Lowcock E, Naus M, Lee V. Effectiveness and cost-effectiveness of pediatric rotavirus vaccination in British Columbia: A model-based evaluation. Vaccine 2012; 30:7601-7. [DOI: 10.1016/j.vaccine.2012.10.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 09/29/2012] [Accepted: 10/10/2012] [Indexed: 10/27/2022]
|
308
|
Guerra AH, Stockmann C, Pavia AT, Hersh AL, Thorell EA, Weng HY, Korgenski K, Byington CL, Ampofo K. Laboratory-Confirmed Rotavirus Disease in Utah Children: Clinical and Economic Impact of Rotavirus Vaccination. J Pediatric Infect Dis Soc 2012; 1:268-77. [PMID: 23687580 PMCID: PMC3656544 DOI: 10.1093/jpids/pis058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2011] [Accepted: 03/27/2012] [Indexed: 11/14/2022]
Abstract
BACKGROUND Rotavirus is the most common cause of infectious diarrhea in children worldwide. Recent studies have described changes in the burden of all-cause gastroenteritis; however, there are limited data on the clinical and economic impact of rotavirus vaccine on cases of laboratory-confirmed rotavirus disease. METHODS We performed a retrospective study of laboratory-confirmed rotavirus disease from July 2003 through June 2010 at a children's hospital and a community hospital in Utah. Demographics and hospital costs for children <5 years with rotavirus symptoms and a positive rotavirus enzyme immunoassay test on a stool specimen were abstracted from electronic medical records. We compared the prevaccine period (2003-2007) with the postvaccine period (2008-2010). RESULTS The overall incidence of rotavirus gastroenteritis declined in the postvaccine period, from 26.6 to 5.2 cases per 10 000 person-years for Salt Lake County residents. The largest decrease in the incidence of rotavirus gastroenteritis was among children <12 months (-87%; 95% confidence interval [CI], 79-93). Older children (12-23 months) also experienced significant decreases (-81%; 95% CI, 72-88), as did those 24-59 months (-61%; 95% CI, 51-71). In 2009, 3 years after rotavirus vaccine introduction, there was a 79% decrease in emergency department visits and a 78% decrease in hospitalizations across both hospitals. The cost of emergency department visits and hospitalizations for rotavirus gastroenteritis decreased by 79% and 72%, respectively, resulting in annual savings of $790 000 at a children's hospital and $140 000 at a community hospital. CONCLUSION Rotavirus vaccination in infants has dramatically decreased the clinical burden and direct medical costs of rotavirus gastroenteritis in both infants and young children.
Collapse
Affiliation(s)
- Angel Herrera Guerra
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Utah Health Sciences Center, Salt Lake City
| | | | - Andrew T. Pavia
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Utah Health Sciences Center, Salt Lake City
| | - Adam L. Hersh
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Utah Health Sciences Center, Salt Lake City
| | - Emily A. Thorell
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Utah Health Sciences Center, Salt Lake City
| | - Hsin Yi Weng
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Utah Health Sciences Center, Salt Lake City
| | - Kent Korgenski
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Utah Health Sciences Center, Salt Lake City
| | - Carrie L. Byington
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Utah Health Sciences Center, Salt Lake City
| | - Krow Ampofo
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Utah Health Sciences Center, Salt Lake City
| |
Collapse
|
309
|
Soares-Weiser K, Maclehose H, Bergman H, Ben-Aharon I, Nagpal S, Goldberg E, Pitan F, Cunliffe N. Vaccines for preventing rotavirus diarrhoea: vaccines in use. Cochrane Database Syst Rev 2012; 11:CD008521. [PMID: 23152260 DOI: 10.1002/14651858.cd008521.pub3] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Rotavirus results in more diarrhoea-related deaths in children less than five years of age than any other single agent in countries with high childhood mortality. It is also a common cause of diarrhoea-related hospital admissions in countries with low childhood mortality. Currently licensed rotavirus vaccines include a monovalent rotavirus vaccine (RV1; Rotarix, GlaxoSmithKline Biologicals) and a pentavalent rotavirus vaccine (RV5; RotaTeq, Merck & Co., Inc.). Lanzhou lamb rotavirus vaccine (LLR; Lanzhou Institute of Biomedical Products) is used in China only. OBJECTIVES To evaluate rotavirus vaccines approved for use (RV1, RV5, and LLR) for preventing rotavirus diarrhoea. SEARCH METHODS We searched MEDLINE (via PubMed) (1966 to May 2012), the Cochrane Infectious Diseases Group Specialized Register (10 May 2012), CENTRAL (published in The Cochrane Library 2012, Issue 5), EMBASE (1974 to 10 May 2012), LILACS (1982 to 10 May 2012), and BIOSIS (1926 to 10 May 2012). We also searched the ICTRP (10 May 2012), www.ClinicalTrials.gov (28 May 2012) and checked reference lists of identified studies. SELECTION CRITERIA We selected randomized controlled trials (RCTs) in children comparing rotavirus vaccines approved for use with placebo, no intervention, or another vaccine. DATA COLLECTION AND ANALYSIS Two authors independently assessed trial eligibility, extracted data, and assessed risk of bias. We combined dichotomous data using the risk ratio (RR) and 95% confidence intervals (CI). We stratified the analysis by child mortality, and used GRADE to evaluate evidence quality. MAIN RESULTS Forty-one trials met the inclusion criteria and enrolled a total of 186,263 participants. Twenty-nine trials (101,671 participants) assessed RV1, and 12 trials (84,592 participants) evaluated RV5. We did not find any trials assessing LLR.RV1Children aged less than one year: In countries with low-mortality rates, RV1 prevents 86% of severe rotavirus diarrhoea cases (RR 0.14, 95% CI 0.07 to 0.26; 40,631 participants, six trials; high-quality evidence), and, based on one large multicentre trial in Latin America and Finland, probably prevents 40% of severe all-cause diarrhoea episodes (rate ratio 0.60, 95% CI 0.50 to 0.72; 17,867 participants, one trial; moderate-quality evidence). In countries with high-mortality rates, RV1 probably prevents 63% of severe rotavirus diarrhoea cases (RR 0.37, 95% CI 0.18 to 0.75; 5414 participants, two trials; moderate-quality evidence), and, based on one trial in Malawi and South Africa, 34% of severe all-cause diarrhoea cases (RR 0.66, 95% CI 0.44 to 0.98; 4939 participants, one trial; moderate-quality evidence).Children aged up to two years: In countries with low-mortality rates, RV1 prevents 85% of severe rotavirus diarrhoea cases (RR 0.15, 95% CI 0.12 to 0.20; 32,854 participants, eight trials; high-quality evidence), and probably 37% of severe all-cause diarrhoea episodes (rate ratio 0.63, 95% CI 0.56 to 0.71; 39,091 participants, two trials; moderate-quality evidence). In countries with high-mortality rates, based on one trial in Malawi and South Africa, RV1 probably prevents 42% of severe rotavirus diarrhoea cases (RR 0.58, 95% CI 0.42 to 0.79; 2764 participants, one trial; moderate-quality evidence), and 18% of severe all-cause diarrhoea cases (RR 0.82, 95% CI 0.71 to 0.95; 2764 participants, one trial; moderate-quality evidence).RV5Children aged less than one year: In countries with low-mortality rates, RV5 probably prevents 87% of severe rotavirus diarrhoea cases (RR 0.13, 95% CI 0.04 to 0.45; 2344 participants, three trials; moderate-quality evidence), and, based on one trial in Finland, may prevent 72% of severe all-cause diarrhoea cases (RR 0.28, 95% CI 0.16 to 0.48; 1029 participants, one trial; low-quality evidence). In countries with high-mortality rates, RV5 prevents 57% of severe rotavirus diarrhoea (RR 0.43, 95% CI 0.29 to 0.62; 5916 participants, two trials; high-quality evidence), but there was insufficient data to assess the effect on severe all-cause diarrhoea.Children aged up to two years: Four studies provided data for severe rotavirus and all-cause diarrhoea in countries with low-mortality rates. Three trials reported on severe rotavirus diarrhoea cases and found that RV5 probably prevents 82% (RR 0.18, 95% CI 0.07 to 0.50; 3190 participants, three trials; moderate-quality evidence), and another trial in Finland reported on severe all-cause diarrhoea cases and found that RV5 may prevent 96% (RR 0.04, 95% CI 0.00 to 0.70; 1029 participants, one trial; low-quality evidence). In high-mortality countries, RV5 prevents 41% of severe rotavirus diarrhoea cases (RR 0.59, 95% CI 0.43 to 0.82; 5885 participants, two trials; high-quality evidence), and 15% of severe all-cause diarrhoea cases (RR 0.85, 95% CI 0.75 to 0.98; 5977 participants, two trials; high-quality evidence).There was no evidence of a vaccine effect on mortality (181,009 participants, 34 trials; low-quality evidence), although the trials were not powered to detect an effect on this end point.Serious adverse events were reported in 4565 out of 99,438 children vaccinated with RV1 and in 1884 out of 78,226 children vaccinated with RV5. Fifty-eight cases of intussusception were reported in 97,246 children after RV1 vaccination, and 34 cases in 81,459 children after RV5 vaccination. No significant difference was found between children receiving RV1 or RV5 and placebo in the number of serious adverse events, and intussusception in particular. AUTHORS' CONCLUSIONS RV1 and RV5 prevent episodes of rotavirus diarrhoea. The vaccine efficacy is lower in high-mortality countries; however, due to the higher burden of disease, the absolute benefit is higher in these settings. No increased risk of serious adverse events including intussusception was detected, but post-introduction surveillance studies are required to detect rare events associated with vaccination.
Collapse
|
310
|
Justino MCA, Araújo EC, van Doorn LJ, Oliveira CS, Gabbay YB, Mascarenhas JDP, Miranda YS, Guerra SDFS, Silva VBD, Linhares AC. Oral live attenuated human rotavirus vaccine (RotarixTM) offers sustained high protection against severe G9P[8] rotavirus gastroenteritis during the first two years of life in Brazilian children. Mem Inst Oswaldo Cruz 2012; 107:846-53. [DOI: 10.1590/s0074-02762012000700002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 07/18/2012] [Indexed: 11/21/2022] Open
|
311
|
Leino T, Ollgren J, Salo H, Tiihonen P, Kilpi T. First year experience of rotavirus immunisation programme in Finland. Vaccine 2012; 31:176-82. [PMID: 23122991 DOI: 10.1016/j.vaccine.2012.10.068] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 10/16/2012] [Accepted: 10/20/2012] [Indexed: 11/26/2022]
Abstract
INTRODUCTION This study aimed to estimate the impact of rotavirus (RV) immunisation programme on the total hospital treated acute gastroenteritis (AGE) burden, as well as, on severe RV disease burden in Finland during the first year after immunisation programme introduction. Such studies can also be considered as a vaccine-probe-study, where unspecific disease burden prevented by immunisation is assumed to be caused by the agent the vaccine is targeted against. METHODS The RV related outcome definitions were based on data registered in the National Hospital Discharge Register coded using ICD 10 codes. Incidences of hospitalised and hospital outpatient cases of AGE and RVGE were compared prior (1999-2005) and after (2010) the start of the programme among children under 5 years of age. ICD 10 codes utilised were A00-A09, R11 and K52. RESULTS The reductions in disease burden, when the post-introduction year was compared to pre-vaccine era, were 80.3% (95% CI 74.5-84.7) in hospital inpatient RVGE among toddlers less than 1 year of age and 53.9% (95% CI 49.8-57.7) when the total inpatient AGE burden was considered in the same age group. For the corresponding hospital outpatient cases the reductions were 78.8% (95% CI 48.4-91.3) and 12.5% (7.1-17.7). The overall vaccine impact against confirmed RVGE in age cohorts eligible for vaccination before the RV season 2010 was 97% (95% CI 90.7-99.0). If the total reductions, both in diagnosed RVGE, as well as in cases without definite microbial diagnosis, were expected to be RVGE, population based estimates for the total disease burden can be obtained: for inpatient RVGE in children less than 1 year of age the estimate is 10.5/1000 pyrs, while the diagnosed specific incidence was less than half of that, 4.9/1000 pyrs. DISCUSSION During the first post-vaccination year 2010, RV immunisation programme clearly managed to control the severe, hospital treated, forms of RVGE. The total disease burden is a more valuable end point than mere diagnosed cases as laboratory confirmation practises change after vaccine introduction. Our study is limited by the very short post-introduction follow up.
Collapse
Affiliation(s)
- Tuija Leino
- Department of Vaccination and Immune Protection, National Institute for Health and Welfare (THL), Mannerheimintie 166, Helsinki, Finland.
| | | | | | | | | |
Collapse
|
312
|
The cost-effectiveness of pentavalent rotavirus vaccination in England and Wales. Vaccine 2012; 30:6766-76. [DOI: 10.1016/j.vaccine.2012.09.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 09/04/2012] [Accepted: 09/10/2012] [Indexed: 11/23/2022]
|
313
|
Lepage P, Vergison A. Impact of rotavirus vaccines on rotavirus disease. Expert Rev Anti Infect Ther 2012; 10:547-61. [PMID: 22702319 DOI: 10.1586/eri.12.39] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Rotaviruses are the most common cause of acute gastroenteritis in young children worldwide. Both licensed rotavirus vaccines (Rotarix™ [RV1] and RotaTeq™ [RV5]) are effective and safe. Studies from countries that have included RV1 or RV5 in the national immunization programs have demonstrated their safety and sustained efficacy under real-life circumstances. A significant decline in acute gastroenteritis-related deaths among Latin American children was observed after the introduction of RV1 and RV5 vaccines. Both vaccines were able to decrease the number of cases of rotavirus acute gastroenteritis and of severe rotavirus diseases. Vaccination was also associated with a dramatic reduction in hospitalizations and outpatient visits for all-cause acute gastroenteritis. Indirect protection after infant mass vaccination has been strongly suggested. Moreover, postlicensure safety studies assessed rare adverse events (rates <1 in 50,000), such as intussusception.
Collapse
Affiliation(s)
- Philippe Lepage
- Université Libre de Bruxelles and Infectious Diseases Unit, Hôpital Universitaire des Enfants Reine Fabiola, Brussels, Belgium.
| | | |
Collapse
|
314
|
Benhafid M, Elomari N, Elqazoui M, Meryem AI, Rguig A, Filali-Maltouf A, Elaouad R. Diversity of rotavirus strains circulating in children under 5 years of age admitted to hospital for acute gastroenteritis in Morocco, June 2006 to May 2009. J Med Virol 2012; 85:354-62. [PMID: 23074038 DOI: 10.1002/jmv.23445] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2012] [Indexed: 01/28/2023]
Abstract
Rotavirus vaccine was introduced in Morocco during 2010. In anticipation of introducing rotavirus vaccines, the Ministry of Health in Morocco established a rotavirus surveillance network in June 2006 at four hospitals in Morocco to obtain baseline data on rotavirus disease burden and prevalent strains. From June 2006 to May 2009, stool samples were collected from children under 5 years of age admitted for diarrhea to four sentinel hospitals serving different regions of Morocco. Rotaviruses were detected in stools using enzyme immunoassay, then genotyped by reverse-transcriptase polymerase chain reaction. Samples with adequate stool in which the P or G types could not be determined by RT-PCR were subjected to nucleotide sequence analysis. Overall, 42% (579 of 1,388) of the stools samples tested were positive for rotavirus. Genotyping of 548 (95%) samples demonstrated that G1P[8] (55%) was the most prevalent strain, followed by G9P[8] (11.3%), G2P[4] (9.1%), G4P[8] (0.9%), and G3P[8] (0.4%). Several other strains were identified including G1P[4] (0.2%), G1P[6] (0.9%), G2P[6] (4.3%), G2P[8] (0.2%), G3P[6] (0.4%), G3P[4] (0.2%), and G9P[6] (0.2%). A high prevalence of mixed infections was found (15% of all samples) of which G1G2P[8] (4%) and G1G3P[8] (3.6%) accounted for the majority. Considerable diversity of rotavirus genotypes was present among strains circulating in Morocco prior to the introduction of the vaccine. This study highlighted the need for maintaining active surveillance to monitor changes in rotavirus disease burden and strain dynamics and to detect changes over time that could impact the effectiveness of the vaccination program.
Collapse
Affiliation(s)
- Mohammed Benhafid
- Virology Laboratory, National Institute of Hygiene, Ministry of Health, Rabat, Morocco.
| | | | | | | | | | | | | |
Collapse
|
315
|
Parez N, Mory O, Pozzetto B, Garbag-Chenon A, Pillet S, Texier N, Téhard B. [Impact of Rotavirus gastroenteritis requiring hospitalization or presenting to emergency room among children less than 5 years in France]. PATHOLOGIE-BIOLOGIE 2012; 60:275-281. [PMID: 21807470 DOI: 10.1016/j.patbio.2011.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Accepted: 04/07/2011] [Indexed: 10/17/2022]
Abstract
BACKGROUND In France, Rotavirus infections are responsible for approximately 300,000 cases of acute gastroenteritis (AGE) in children less than 5 years every year, 138,000 outpatient consultations and 18,000 hospitalizations. Surveillance for Rotavirus Infections in Hospitalized Kids (SHRIK) is a European prospective observational study conducted to assess the burden related to Rotavirus gastroenteritis (RVGE) in these children. METHODS Patients less than 5 years visiting emergency rooms for AGE (U-AGE), who were hospitalized (H-AGE) or who developed RVGE 48 hours after hospitalization (N-AGE) were included in the study over a year. A stool sample was collected for every child and analyzed by ELISA. RESULTS Results are presented for patients enrolled in France. A total of 755 eligible patients with AGE were included (357 for U-AGE, 372 for H-AGE and 26 for N-AGE). Among them, the proportion of RVGE was 49.1% (n=114) for U-AGE and 64.4% (n=186) for H-AGE. Most cases of RVGE (89%) involved children less than 2 years. GERV were frequently more severe than GEA non related to Rotavirus (NRVGE), according to the Vesikari scale, (68.4% against 41.9%, P<0.0001). Oral rehydration was performed for nearly 50% of RVGE patients before coming to hospital, versus 36.2% for NRVGE (P<0.002). All RV-positive strains were genotyped: the most frequent strains were G1P[8] (U-AGE, 42%; H-AGE, 46%) and G9P[8] (U-AGE, 38%; H-AGE, 31%). CONCLUSION SHRIK study followed up all GEA visiting emergency room or requiring hospitalization for one year and showed that the burden of Rotavirus disease is high with a ratio over 70% of all hospital GEA during the winter peak.
Collapse
Affiliation(s)
- N Parez
- Hôpital Louis-Mourier, 178, rue des Renouillers, 92701 Colombes cedex, France.
| | | | | | | | | | | | | |
Collapse
|
316
|
Williams DJ, Edwards KM, Payne DC, Manning J, Parashar UD, Lopman BA. Decline in gastroenteritis-related triage calls after rotavirus vaccine licensure. Pediatrics 2012; 130:e872-8. [PMID: 22966021 DOI: 10.1542/peds.2012-0330] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE We sought to determine the impact of rotavirus vaccine implementation on gastroenteritis (GE)-related calls to a large telephone triage service in Tennessee. METHODS Total and GE-related calls received by the Vanderbilt Telephone Triage Program for children <5 years of age were examined from May 1, 2004 to April 30, 2010. Time series adapted Poisson regression models were used to compare weekly GE-related call proportions between the prevaccine (May 2004 to April 2007) and postlicensure (May 2007 to April 2010) periods. Separate models compared GE-related call proportions in the historical rotavirus (February to April) and nonrotavirus (May to January) seasons. Associations between call data and laboratory-confirmed rotavirus detections and regionally reported norovirus activity were also assessed. RESULTS There were 156362 total calls and 19731 GE-related calls. Annual GE-related call proportions declined by 8% (95% confidence interval, 3%-12%) in the postlicensure period; declines ranging from 23% to 31% occurred during the historical rotavirus season in all 3 postlicensure years. No declines occurred in the nonrotavirus season. After vaccine licensure, reductions in laboratory-confirmed rotavirus activity were associated with declines in GE-related call proportions. Peak GE-related call proportions in the postlicensure period occurred earlier than in prevaccine years and were not strongly associated with laboratory-confirmed rotavirus but instead showed good correlation with norovirus outbreaks. CONCLUSIONS A decline in GE-related call proportions among young children after rotavirus vaccine licensure was documented by using a novel surveillance platform that captures mild GE not detected in other surveillance systems. Since rotavirus vaccine licensure, peak call proportions correlate with regional norovirus activity, highlighting the role of that pathogen in community GE.
Collapse
Affiliation(s)
- Derek J Williams
- Division of Hospital Medicine, Department of Pediatrics, Vanderbilt Vaccine Research Program, Vanderbilt University School of Medicine and the Monroe Carell, Jr. Children’s Hospital at Vanderbilt, Nashville, Tennessee, USA.
| | | | | | | | | | | |
Collapse
|
317
|
Steele AD, Neuzil KM, Cunliffe NA, Madhi SA, Bos P, Ngwira B, Witte D, Todd S, Louw C, Kirsten M, Aspinall S, Van Doorn LJ, Bouckenooghe A, Suryakiran PV, Han HH. Human rotavirus vaccine Rotarix™ provides protection against diverse circulating rotavirus strains in African infants: a randomized controlled trial. BMC Infect Dis 2012; 12:213. [PMID: 22974466 PMCID: PMC3462149 DOI: 10.1186/1471-2334-12-213] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 08/22/2012] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Rotaviruses are the most important cause of severe acute gastroenteritis worldwide in children <5 years of age. The human, G1P[8] rotavirus vaccine Rotarix™ significantly reduced severe rotavirus gastroenteritis episodes in a Phase III clinical trial conducted in infants in South Africa and Malawi. This paper examines rotavirus vaccine efficacy in preventing severe rotavirus gastroenteritis, during infancy, caused by the various G and P rotavirus types encountered during the first rotavirus-season. METHODS Healthy infants aged 5-10 weeks were enrolled and randomized into three groups to receive either two (10 and 14 weeks) or three doses of Rotarix™ (together forming the pooled Rotarix™ group) or three doses of placebo at a 6,10,14-week schedule. Weekly home visits were conducted to identify gastroenteritis episodes. Rotaviruses were detected by ELISA and genotyped by RT-PCR and nucleotide sequencing. The percentage of infants with severe rotavirus gastroenteritis caused by the circulating G and P types from 2 weeks post-last dose until one year of age and the corresponding vaccine efficacy was calculated with 95% CI. RESULTS Overall, 4939 infants were vaccinated and 4417 (pooled Rotarix™ = 2974; placebo = 1443) were included in the per protocol efficacy cohort. G1 wild-type was detected in 23 (1.6%) severe rotavirus gastroenteritis episodes from the placebo group. This was followed in order of detection by G12 (15 [1%] in placebo) and G8 types (15 [1%] in placebo). Vaccine efficacy against G1 wild-type, G12 and G8 types were 64.1% (95% CI: 29.9%; 82%), 51.5% (95% CI:-6.5%; 77.9%) and 64.4% (95% CI: 17.1%; 85.2%), respectively. Genotype P[8] was the predominant circulating P type and was detected in 38 (2.6%) severe rotavirus gastroenteritis cases in placebo group. The remaining circulating P types comprised of P[4] (20 [1.4%] in placebo) and P[6] (13 [0.9%] in placebo). Vaccine efficacy against P[8] was 59.1% (95% CI: 32.8%; 75.3%), P[4] was 70.9% (95% CI: 37.5%; 87.0%) and P[6] was 55.2% (95% CI: -6.5%; 81.3%) CONCLUSIONS Rotarix™ vaccine demonstrated efficacy against severe gastroenteritis caused by diverse circulating rotavirus types. These data add to a growing body of evidence supporting heterotypic protection provided by Rotarix™. TRIAL REGISTRATION NUMBER NCT00241644.
Collapse
Affiliation(s)
- Andrew Duncan Steele
- Rotavirus Vaccine Program, PATH, 2201 Westlake Ave, Seattle, WA, 98121, USA
- Initiative for Vaccine Research, WHO, 22 Appia Ave, Geneva, 1211, Switzerland
| | - Kathleen M Neuzil
- Rotavirus Vaccine Program, PATH, 2201 Westlake Ave, Seattle, WA, 98121, USA
| | - Nigel A Cunliffe
- Department of Clinical Infection, Microbiology & Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, England
| | - Shabir A Madhi
- National Institute for Communicable Diseases: a division of National Health Laboratory Services, Sandringham, South Africa
- Department of Science and Technology National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | - Pieter Bos
- MRC Diarrhoeal Pathogens Research Unit, University of Limpopo, Limpopo, South Africa
| | - Bagrey Ngwira
- Department of Community Health, College of Medicine, Blantyre, Malawi
| | - Desiree Witte
- Department of Clinical Infection, Microbiology & Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, England
- Department of Community Health, College of Medicine, Blantyre, Malawi
| | - Stacy Todd
- Department of Clinical Infection, Microbiology & Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, England
| | - Cheryl Louw
- Madibeng Centre for Research, Brits, South Africa
| | - Mari Kirsten
- Department of Paediatric Surgery, University of Pretoria, Pretoria, RSA
| | - Sanet Aspinall
- Synexus Clinical Research SA/Rota Consortium, Pretoria, South Africa
| | | | | | | | | |
Collapse
|
318
|
Fulfilling the promise of rotavirus vaccines: how far have we come since licensure? THE LANCET. INFECTIOUS DISEASES 2012; 12:561-70. [PMID: 22742639 DOI: 10.1016/s1473-3099(12)70029-4] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Rotavirus is the most common cause of fatal and severe childhood diarrhoea worldwide. Two new rotavirus vaccines have shown efficacy against severe rotavirus disease in large clinical trials. Between 2006 and 2010, 27 countries introduced rotavirus vaccination into national immunisation programmes and, subsequently, the burden of severe rotavirus disease in these countries has decreased substantially in both vaccinated and unvaccinated children. Rotavirus vaccination has led to large, sustained declines in childhood deaths from diarrhoea in Brazil and Mexico, which supports estimates that rotavirus was the leading cause of diarrhoeal deaths in these countries. Studies after licensing have provided new insights into these vaccines, such as the duration of protection, relative effectiveness in poor populations, and strain evolution after vaccine introduction. The challenge for policy makers worldwide is to analyse the effect of vaccination in early adopter countries and to assess whether the benefits outweigh the costs and encourage wider dissemination of these vaccines.
Collapse
|
319
|
Dudareva-Vizule S, Koch J, An der Heiden M, Oberle D, Keller-Stanislawski B, Wichmann O. Impact of rotavirus vaccination in regions with low and moderate vaccine uptake in Germany. Hum Vaccin Immunother 2012; 8:1407-15. [PMID: 22960495 DOI: 10.4161/hv.21593] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In Germany, routine RV-vaccination is not adopted into the national immunization schedule as of 2012. Because RV-vaccines were already on the market since 2006, in 2010 a moderate (58%) and low (22%) vaccine uptake was observed in the 5 eastern federal states (EFS) and the 11 western federal states (WFS), respectively. To assess the impact of RV-vaccination, we compared the incidence rates (IR) of RV-related hospitalizations before (2004‒2006) and in seasons after (2008/09-2010/11) RV-vaccine introduction in Germany by utilizing data from the national mandatory disease reporting system. In the EFS, the IR was significantly reduced in age-groups < 18 mo in 2008/09 and in age-groups < 24 mo in 2009/10-2010/11. In the WFS an IR-reduction was observed only in age-groups < 12 mo in 2008/09 and in age-groups < 18 mo in 2009/10-2010/11. Overall IR-reduction in age-groups < 24 mo comparing 2008-11 with 2004-06 was 36% and 25% in EFS and WFS, respectively. In addition, we computed IR-ratios (IRR) in the seasons after mid-2006 with negative binomial regression. The effect of vaccination was independent from the geographic region. Vaccination was associated with a significant reduction in RV-related hospitalizations in the age-groups 6-23 mo. Most prominently, vaccination of 50% of infants led to an estimated decrease in age group 6-11 mo by 42%. No significant reduction was observed in age-groups ≥ 24 mo. In conclusion, in the German setting with low to moderate vaccine uptake, RV-related hospitalization incidence decreased substantially depending on the achieved vaccination coverage, but only in the first two years of life.
Collapse
|
320
|
Pitzer VE, Atkins KE, de Blasio BF, Van Effelterre T, Atchison CJ, Harris JP, Shim E, Galvani AP, Edmunds WJ, Viboud C, Patel MM, Grenfell BT, Parashar UD, Lopman BA. Direct and indirect effects of rotavirus vaccination: comparing predictions from transmission dynamic models. PLoS One 2012; 7:e42320. [PMID: 22912699 PMCID: PMC3418263 DOI: 10.1371/journal.pone.0042320] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 07/03/2012] [Indexed: 11/25/2022] Open
Abstract
Early observations from countries that have introduced rotavirus vaccination suggest that there may be indirect protection for unvaccinated individuals, but it is unclear whether these benefits will extend to the long term. Transmission dynamic models have attempted to quantify the indirect protection that might be expected from rotavirus vaccination in developed countries, but results have varied. To better understand the magnitude and sources of variability in model projections, we undertook a comparative analysis of transmission dynamic models for rotavirus. We fit five models to reported rotavirus gastroenteritis (RVGE) data from England and Wales, and evaluated outcomes for short- and long-term vaccination effects. All of our models reproduced the important features of rotavirus epidemics in England and Wales. Models predicted that during the initial year after vaccine introduction, incidence of severe RVGE would be reduced 1.8–2.9 times more than expected from the direct effects of the vaccine alone (28–50% at 90% coverage), but over a 5-year period following vaccine introduction severe RVGE would be reduced only by 1.1–1.7 times more than expected from the direct effects (54–90% at 90% coverage). Projections for the long-term reduction of severe RVGE ranged from a 55% reduction at full coverage to elimination with at least 80% coverage. Our models predicted short-term reductions in the incidence of RVGE that exceeded estimates of the direct effects, consistent with observations from the United States and other countries. Some of the models predicted that the short-term indirect benefits may be offset by a partial shifting of the burden of RVGE to older unvaccinated individuals. Nonetheless, even when such a shift occurs, the overall reduction in severe RVGE is considerable. Discrepancies among model predictions reflect uncertainties about age variation in the risk and reporting of RVGE, and the duration of natural and vaccine-induced immunity, highlighting important questions for future research.
Collapse
Affiliation(s)
- Virginia E. Pitzer
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, United States of America
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Katherine E. Atkins
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Birgitte Freiesleben de Blasio
- Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Infectious Diseases Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | | | - Christina J. Atchison
- Infectious Diseases Epidemiology Unit, Department of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John P. Harris
- Centre for Infections, Department of Gastrointestinal, Emerging and Zoonotic Infections, Health Protection Agency, London, United Kingdom
| | - Eunha Shim
- Deparment of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Alison P. Galvani
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - W. John Edmunds
- Infectious Diseases Epidemiology Unit, Department of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Cécile Viboud
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Manish M. Patel
- Epidemiology Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Bryan T. Grenfell
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, United States of America
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Umesh D. Parashar
- Epidemiology Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ben A. Lopman
- Epidemiology Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| |
Collapse
|
321
|
Braeckman T, Van Herck K, Meyer N, Pirçon JY, Soriano-Gabarró M, Heylen E, Zeller M, Azou M, Capiau H, De Koster J, Maernoudt AS, Raes M, Verdonck L, Verghote M, Vergison A, Matthijnssens J, Van Ranst M, Van Damme P. Effectiveness of rotavirus vaccination in prevention of hospital admissions for rotavirus gastroenteritis among young children in Belgium: case-control study. BMJ 2012; 345:e4752. [PMID: 22875947 PMCID: PMC3414434 DOI: 10.1136/bmj.e4752] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To evaluate the effectiveness of rotavirus vaccination among young children in Belgium. DESIGN Prospective case-control study. SETTING Random sample of 39 Belgian hospitals, February 2008 to June 2010. PARTICIPANTS 215 children admitted to hospital with rotavirus gastroenteritis confirmed by polymerase chain reaction and 276 age and hospital matched controls. All children were of an eligible age to have received rotavirus vaccination (that is, born after 1 October 2006 and aged ≥ 14 weeks). MAIN OUTCOME MEASURE Vaccination status of children admitted to hospital with rotavirus gastroenteritis and matched controls. RESULTS 99 children (48%) admitted with rotavirus gastroenteritis and 244 (91%) controls had received at least one dose of any rotavirus vaccine (P<0.001). The monovalent rotavirus vaccine accounted for 92% (n=594) of all rotavirus vaccine doses. With hospital admission as the outcome, the unadjusted effectiveness of two doses of the monovalent rotavirus vaccine was 90% (95% confidence interval 81% to 95%) overall, 91% (75% to 97%) in children aged 3-11 months, and 90% (76% to 96%) in those aged ≥ 12 months. The G2P[4] genotype accounted for 52% of cases confirmed by polymerase chain reaction with eligible matched controls. Vaccine effectiveness was 85% (64% to 94%) against G2P[4] and 95% (78% to 99%) against G1P[8]. In 25% of cases confirmed by polymerase chain reaction with eligible matched controls, there was reported co-infection with adenovirus, astrovirus and/or norovirus. Vaccine effectiveness against co-infected cases was 86% (52% to 96%). Effectiveness of at least one dose of any rotavirus vaccine (intention to vaccinate analysis) was 91% (82% to 95%). CONCLUSIONS Rotavirus vaccination is effective for the prevention of admission to hospital for rotavirus gastroenteritis among young children in Belgium, despite the high prevalence of G2P[4] and viral co-infection.
Collapse
Affiliation(s)
- Tessa Braeckman
- Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
322
|
Donato CM, Cannan D, Bogdanovic-Sakran N, Snelling TL, Kirkwood CD. Characterisation of a G9P[8] rotavirus strain identified during a gastroenteritis outbreak in Alice Springs, Australia post Rotarix™ vaccine introduction. Vaccine 2012; 30 Suppl 1:A152-8. [PMID: 22520125 DOI: 10.1016/j.vaccine.2011.07.119] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 07/19/2011] [Accepted: 07/25/2011] [Indexed: 12/31/2022]
Abstract
A large rotavirus gastroenteritis outbreak occurred in the Alice Springs region of the Northern Territory, Australia from the 12th of March until the 11th of July 2007. The outbreak occurred five months after the introduction of the Rotarix™ vaccine. Electropherotype and sequence analysis demonstrated that a single G9P[8] strain was responsible for the outbreak and that the strain remained highly conserved during the outbreak period. The outbreak strain contained amino acid changes in regions of the VP7 and NSP4 genes, with known biological function, when compared to previously characterised G9P[8] strains from Australia and other international locations. The recent vaccine introduction was unlikely to have influenced genotype selection in this setting. Importantly, Rotarix™ vaccine was highly effective against the G9P[8] outbreak strain.
Collapse
Affiliation(s)
- C M Donato
- Enteric Virus Group, Murdoch Childrens Research Institute, Royal Children's Hospital, Flemington Road, Parkville, Melbourne, VIC 3052, Australia
| | | | | | | | | |
Collapse
|
323
|
Bányai K, László B, Duque J, Steele AD, Nelson EAS, Gentsch JR, Parashar UD. Systematic review of regional and temporal trends in global rotavirus strain diversity in the pre rotavirus vaccine era: insights for understanding the impact of rotavirus vaccination programs. Vaccine 2012; 30 Suppl 1:A122-30. [PMID: 22520121 DOI: 10.1016/j.vaccine.2011.09.111] [Citation(s) in RCA: 347] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 08/30/2011] [Accepted: 09/23/2011] [Indexed: 01/08/2023]
Abstract
Recently, two rotavirus vaccines have been recommended for routine immunization of infants worldwide. These vaccines proved efficacious during clinical trials and field use in both developing and developed countries, and appear to provide good protection against a range of rotavirus genotypes, including some that are not included in the vaccines. However, since conclusive data that the vaccines will protect against a wide variety of rotavirus strains are still lacking and since vaccines may exert some selection pressure, a detailed picture of global strain prevalence from the pre-rotavirus vaccine era is important to evaluate any potential changes in circulating strains observed after widespread introduction of rotavirus vaccines. Thus, we systematically reviewed rotavirus genotyping studies spanning a 12-year period from 1996 to 2007. In total, ~110,000 strains were genotyped from 100 reporting countries. Five genotypes (G1-G4, and G9) accounted for 88% of all strains, although extensive geographic and temporal differences were observed. For example, the prevalence of G1 strains declined from 2000 onward, while G3 strains re-emerged, and G9 and G12 strains emerged during the same period. When crude strain prevalence data were weighted by region based on the region's contribution to global rotavirus mortality, the importance of genotypes G1 and G9 strains that were more prevalent in regions with low mortality was reduced and conversely the importance of G8 strains that were more prevalent in African settings with greater contribution to global rotavirus mortality was increased. This study provides the most comprehensive, up-to-date information on rotavirus strain surveillance in the pre-rotavirus vaccine era and will provide useful background to examine the impact of rotavirus vaccine introduction on future strain prevalence.
Collapse
Affiliation(s)
- Krisztián Bányai
- Veterinary Medical Research Institute, Hungarian Academy of Sciences, Budapest, Hungary.
| | | | | | | | | | | | | |
Collapse
|
324
|
Abstract
Live attenuated oral rotavirus vaccines were tested for proof-of-concept in the early 1980s, the first vaccine (RotaShield, Wyeth) was introduced in 1998 but was subsequently withdrawn because of association with intussusception, and the two currently licensed vaccine (Rotarix, GlaxoSmithKline, and RotaTeq, Merck) were introduced in 2006. Before licensure both vaccines were extensively tested for safety (for intussusception) and efficacy in trials comprising in over 60,000 infants each. Rotarix is a single-strain human rotavirus vaccine (RV1) and RotaTeq is a combination of five bovine-human reassortant rotaviruses (RV5). Although the composition of the two vaccines is different, their field effectiveness and, largely, mechanism of action are similar. Both prevent effectively severe rotavirus gastroenteritis (RVGE) but are less efficacious against mild RVGE or rotavirus infection. Field effectiveness of these vaccines in Europe and the USA against severe RVGE has been above 90% and in Latin America around 80%. Trials in Africa have yielded efficacy rates between 50 and 80%. Rotavirus vaccination has been introduced into the national immunization programmes of about 20 countries in Latin America, with Brazil and Mexico as leading countries, as well as in the USA, Australia and South Africa. Introduction into other African countries will start in 2012. In Europe, Belgium, Luxembourg, Austria and Finland and five federal states of Germany have introduced universal rotavirus vaccination. The reasons for the slow progress in Europe include low mortality from RVGE, unfavourable cost-benefit calculations in some countries, and concerns that still exist over intussusception.
Collapse
Affiliation(s)
- T Vesikari
- University of Tampere, Vaccine Research Centre, Tampere, Finland.
| |
Collapse
|
325
|
Lopman BA, Payne DC, Tate JE, Patel MM, Cortese MM, Parashar UD. Post-licensure experience with rotavirus vaccination in high and middle income countries; 2006 to 2011. Curr Opin Virol 2012; 2:434-42. [DOI: 10.1016/j.coviro.2012.05.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 05/01/2012] [Accepted: 05/09/2012] [Indexed: 12/23/2022]
|
326
|
Bucardo F, Rippinger CM, Svensson L, Patton JT. Vaccine-derived NSP2 segment in rotaviruses from vaccinated children with gastroenteritis in Nicaragua. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2012; 12:1282-94. [PMID: 22487061 PMCID: PMC3372771 DOI: 10.1016/j.meegid.2012.03.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Revised: 03/09/2012] [Accepted: 03/10/2012] [Indexed: 10/28/2022]
Abstract
Rotavirus (RV) vaccination programs have been established in several countries using the human-attenuated G1P[8] monovalent vaccine Rotarix (GlaxoSmithKline) and/or the human-bovine reassortant G1, G2, G3, G4, P[8] pentavalent vaccine RotaTeq (Merck). The efficacy of both vaccines is high (∼90%) in developed countries, but can be remarkably lower in developing countries. For example, a vaccine efficacy against severe diarrhea of only 58% was observed in a 2007-2009 Nicaraguan study using RotaTeq. To gain insight into the significant level of vaccine failure in this country, we sequenced the genomes of RVs recovered from vaccinated Nicaraguan children with gastroenteritis. The results revealed that all had genotype specificities typical for human RVs (11 G1P[8], 1 G3P[8]) and that the sequences and antigenic epitopes of the outer capsid proteins (VP4 and VP7) of these viruses were similar to those reported for RVs isolated elsewhere in the world. As expected, nine of the G1P[8] viruses and the single G3P[8] virus had genome constellations typical of human G1P[8] and G3P[8] RVs: G1/3-P[8]-I1-R1-C1-M1-A1-N1-T1-E1-H1. However, two of the G1P[8] viruses had atypical constellations, G1-P[8]-I1-R1-C1-M1-A1-N2-T1-E1-H1, due to the presence of a genotype-2 NSP2 (N2) gene. The sequence of the N2 NSP2 gene was identical to the bovine N2 NSP2 gene of RotaTeq, indicating that the two atypical viruses originated via reassortment of human G1P[8] RVs with RotaTeq viruses. Together, our data suggest that the high level of vaccine failure in Nicaraguan is probably not due to antigenic drift of commonly circulating virus strains nor the emergence of new antigenetically distinct virus strains. Furthermore, our data suggest that the widespread use of the RotaTeq vaccine has led to the introduction of vaccine genes into circulating human RVs.
Collapse
Affiliation(s)
- Filemón Bucardo
- Department of Microbiology, University of León, UNAN-León, Nicaragua
| | - Christine M. Rippinger
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lennart Svensson
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Sweden
| | - John T. Patton
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
327
|
Connolly MP, Topachevskyi O, Standaert B, Ortega O, Postma M. The impact of rotavirus vaccination on discounted net tax revenue in Egypt: a government perspective analysis. PHARMACOECONOMICS 2012; 30:681-95. [PMID: 22788259 DOI: 10.2165/11597750-000000000-00000] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND We evaluated national rotavirus (RV) immunization programme costs to estimate how resulting changes in morbidity and mortality will influence government fiscal accounts over time. The assumption was that increased childhood survival in vaccinated cohorts leads to increased numbers of children consuming government resource, and an increased number of future tax payers. OBJECTIVE Our objective was to evaluate the difference in lifetime discounted net tax revenue generated by RV vaccinated and unvaccinated cohorts from the Egyptian government perspective. METHODS The model framework adopts the Egyptian government perspective for RV immunization costs (year 2009 values) and all government transfers (e.g. education costs, health costs, pensions). To reflect the government tax revenue, we applied a fixed income tax burden to earnings over the lifetime of vaccinated and unvaccinated cohorts. At each year of the model, we derive net taxes (gross taxes less transfers) discounted to the immunization year to reflect the present value of RV vaccination investment costs. RESULTS Projected incremental net present values of the vaccinated cohort versus the unvaccinated cohort are $US6.1 million, $US58.1 million and $US55.7 million at 25-, 50- and 72-year time horizons, respectively. The internal rate of return for the government based on RV vaccination at years 25, 50 and 72 was 10.8%, 15.1% and 14.9, respectively. Within the first 5 years of vaccination, 76% of vaccine acquisition costs were offset due to direct and indirect cost savings attributed to a reduction in RV-related disease burden. Investments in RV vaccination in a single year are entirely offset when the vaccinated cohort of newborns reach 22 years of age. CONCLUSION The government perspective is useful for evaluating investments in RV vaccination because of ongoing government transfers and tax receipts attributed to changes in RV-attributed morbidity and mortality. The analysis described here illustrates that investing in RV offers tangible long-term fiscal benefits for government over many generations that would not ordinarily be captured in economic evaluations typically applied to healthcare interventions.
Collapse
Affiliation(s)
- Mark P Connolly
- PharmacoEpidemiology and PharmacoEconomics, Department of Pharmacy, University of Groningen, Groningen, the Netherlands.
| | | | | | | | | |
Collapse
|
328
|
Dulgheroff ACB, Figueiredo EF, Moreira LP, Moreira KC, Moura LMS, Gouvêa VS, Domingues ALS. Distribution of rotavirus genotypes after vaccine introduction in the Triângulo Mineiro region of Brazil: 4-Year follow-up study. J Clin Virol 2012; 55:67-71. [PMID: 22795600 DOI: 10.1016/j.jcv.2012.06.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 06/09/2012] [Accepted: 06/11/2012] [Indexed: 11/17/2022]
Abstract
BACKGROUND Rotaviruses are the major cause of diarrhea in children for which a monovalent G1P[8] vaccine has been provided free for all Brazilian infants since March 2006. OBJECTIVES To investigate prevalence and genotypes of rotavirus strains causing diarrhea in children in Triângulo Mineiro, Minas Gerais, during 2007-2010, and to assess local vaccine impact. STUDY DESIGN Fecal specimens were analyzed for rotavirus detection and characterization by PAGE, RT-PCR and PCR-genotyping assays. RESULTS Overall, rotavirus was diagnosed in 12.1% (76/630) cases, accounting for 35.8% of the hospitalizations and 6.5% of outpatient attendance due to diarrhea. A trend in rotavirus disease reduction occurred in both cities (71.8% and 83.4% in Uberaba; 95.3% in Uberlândia) up to 2009, but it reversed in 2010 with increased rotavirus cases in Uberlândia. Short pattern G2P[4] strains were detected in all but three (96%) cases of mixed/P[NT] infections with long electropherotypes. CONCLUSIONS This 4-year follow-up study showed a reduction in rotavirus-related diarrhea and even skipped a rotavirus season, which is consistent with vaccine mediated protection. The 2007-2010 rotavirus epidemic curve reflected the natural cyclic fluctuation of the single G2P[4] genotype, with sharp reduction of cases in 2008 leading to lack of a rotavirus 2009 season (both cases and hospitalizations) followed by its come back in 2010. Diarrhea cases related to either vaccine serotype/genotype (G1 or P[8]) were not detected. Thus, a new scenario emerged with a single epidemic genotype replacing the cocirculation of great diversity of genotypes, thus far, a hallmark of the epidemiology of rotavirus in Brazil.
Collapse
Affiliation(s)
- A C B Dulgheroff
- Instituto de Ciências Biológicas e Naturais, Microbiologia, Universidade Federal do Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | | | | | | | | | | | | |
Collapse
|
329
|
Kahn G, Fitzwater S, Tate J, Kang G, Ganguly N, Nair G, Steele D, Arora R, Chawlasarkar M, Parashar U, Santosham M. Epidemiology and prospects for prevention of rotavirus disease in India. Indian Pediatr 2012; 49:467-74. [DOI: 10.1007/s13312-012-0076-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
330
|
Mosca A, Leblanc A, Bellaïche M, Tounian P, Cohen R. [How many deaths before rotavirus vaccination becomes routine?]. Arch Pediatr 2012; 19:783-5. [PMID: 22748690 DOI: 10.1016/j.arcped.2012.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 05/29/2012] [Indexed: 10/26/2022]
|
331
|
Postmarketing surveillance of intussusception following mass introduction of the attenuated human rotavirus vaccine in Mexico. Pediatr Infect Dis J 2012; 31:736-44. [PMID: 22695189 DOI: 10.1097/inf.0b013e318253add3] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Mexico initiated mass vaccination with the attenuated human rotavirus vaccine (Rotarix) in 2006. This postlicensure study aimed to assess any potential temporal association between vaccination and intussusception in Mexican infants. METHODS Prospective, active surveillance for intussusception among infants aged less than 1 year was conducted in 221 hospitals across Mexico from the Mexican Institute of Social Security between January 2008 and October 2010. The temporal association between vaccination and intussusception was assessed by self-controlled case-series analysis. RESULTS Of the 753 episodes of intussusception reported in 750 infants, 701 were in vaccinated infants (34.5% post-dose 1, 65.5% post-dose 2). The relative incidence of intussusception within 31 days of vaccination was 1.75 (95.5% confidence interval [CI]: 1.24-2.48; P=0.001) post-dose 1 and 1.06 (95.5% CI: 0.75-1.48; P=0.75) post-dose 2. The relative incidence of intussusception within 7 days of vaccination was 6.49 post-dose 1 (95.5% CI: 4.17-10.09; P<0.001) and 1.29 post-dose 2 (95.5% CI: 0.80-2.11; P=0.29). Clustering of intussusception within 7 days of vaccination was observed post-dose 1. An attributable risk of 3 to 4 additional cases of intussusception per 100,000 vaccinated infants was estimated. CONCLUSION This is the largest surveillance study for intussusception after rotavirus vaccination to date. A temporal increase in the risk for intussusception was seen within 7 days of administration of the first vaccine dose. It is still uncertain whether rotavirus vaccination has any impact on the overall incidence of intussusception. This finding has to be put in perspective with the well-documented substantial benefits of rotavirus vaccination.
Collapse
|
332
|
Diversity and relationships of cocirculating modern human rotaviruses revealed using large-scale comparative genomics. J Virol 2012; 86:9148-62. [PMID: 22696651 DOI: 10.1128/jvi.01105-12] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Group A rotaviruses (RVs) are 11-segmented, double-stranded RNA viruses and are primary causes of gastroenteritis in young children. Despite their medical relevance, the genetic diversity of modern human RVs is poorly understood, and the impact of vaccine use on circulating strains remains unknown. In this study, we report the complete genome sequence analysis of 58 RVs isolated from children with severe diarrhea and/or vomiting at Vanderbilt University Medical Center (VUMC) in Nashville, TN, during the years spanning community vaccine implementation (2005 to 2009). The RVs analyzed include 36 G1P[8], 18 G3P[8], and 4 G12P[8] Wa-like genogroup 1 strains with VP6-VP1-VP2-VP3-NSP1-NSP2-NSP3-NSP4-NSP5/6 genotype constellations of I1-R1-C1-M1-A1-N1-T1-E1-H1. By constructing phylogenetic trees, we identified 2 to 5 subgenotype alleles for each gene. The results show evidence of intragenogroup gene reassortment among the cocirculating strains. However, several isolates from different seasons maintained identical allele constellations, consistent with the notion that certain RV clades persisted in the community. By comparing the genes of VUMC RVs to those of other archival and contemporary RV strains for which sequences are available, we defined phylogenetic lineages and verified that the diversity of the strains analyzed in this study reflects that seen in other regions of the world. Importantly, the VP4 and VP7 proteins encoded by VUMC RVs and other contemporary strains show amino acid changes in or near neutralization domains, which might reflect antigenic drift of the virus. Thus, this large-scale, comparative genomic study of modern human RVs provides significant insight into how this pathogen evolves during its spread in the community.
Collapse
|
333
|
Matthijnssens J, Van Ranst M. Genotype constellation and evolution of group A rotaviruses infecting humans. Curr Opin Virol 2012; 2:426-33. [PMID: 22683209 DOI: 10.1016/j.coviro.2012.04.007] [Citation(s) in RCA: 235] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 03/01/2012] [Accepted: 04/19/2012] [Indexed: 12/12/2022]
Abstract
Numerous rotavirus group A (RVA) strains with distinct G-genotype and P-genotype combinations have been described infecting humans worldwide. However, the increasing amount of complete RVA genome data which have become available, suggest that only RVA strains with 2 discrete genotype constellations have been successful in sustaining infection of humans worldwide over longer periods of time. Those genotype constellations have been designated I1-R1-C1-M1-A1-N1-T1-E1-H1 and I2-R2-C2-M2-A2-N2-T2-E2-H2 and are also known as Wa-like and DS-1-like, respectively. RVAs of other genotype constellations which were able to spread to a limited extent in the human population are AU-1-related RVA strains (I3-R3-C3-M3-A3/A12-N3-T3-E3-H3/H6) in combination with G3P[9] or G12P[9], and neonatal G10P[11] RVA strains in India (bovine×human Wa-like reassortants). On the basis of the analysis of complete genomes, it is suggested that the overall genetic diversity of epidemiologically widespread human RVA strains is more limited than generally assumed. This conclusion has consequences for how we look at host range restriction and the criteria according to which the effectiveness of RVA universal mass vaccination programs is assessed.
Collapse
Affiliation(s)
- Jelle Matthijnssens
- Laboratory of Clinical & Epidemiological Virology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium.
| | | |
Collapse
|
334
|
Angel J, Franco MA, Greenberg HB. Rotavirus immune responses and correlates of protection. Curr Opin Virol 2012; 2:419-25. [PMID: 22677178 DOI: 10.1016/j.coviro.2012.05.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 05/01/2012] [Accepted: 05/09/2012] [Indexed: 12/11/2022]
Abstract
Selected topics in the field of rotavirus immunity are reviewed focusing on recent developments that may improve efficacy and safety of current and future vaccines. Rotaviruses (RVs) have developed multiple mechanisms to evade interferon (IFN)-mediated innate immunity. Compared to more developed regions of the world, protection induced by natural infection and vaccination is reduced in developing countries where, among other factors, high viral challenge loads are common and where infants are infected at an early age. Studies in developing countries indicate that rotavirus-specific serum IgA levels are not an optimal correlate of protection following vaccination, and better correlates need to be identified. Protection against rotavirus following vaccination is substantially heterotypic; nonetheless, a role for homotypic immunity in selection of circulating postvaccination strains needs further study.
Collapse
Affiliation(s)
- Juana Angel
- Instituto de Genética Humana, Pontificia Universidad Javeriana, Bogotá, Colombia.
| | | | | |
Collapse
|
335
|
Tate JE, Patel MM, Cortese MM, Lopman BA, Gentsch JR, Fleming J, Steele AD, Parashar UD. Remaining issues and challenges for rotavirus vaccine in preventing global childhood diarrheal morbidity and mortality. Expert Rev Vaccines 2012; 11:211-20. [PMID: 22309669 DOI: 10.1586/erv.11.184] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Rotavirus vaccines have had a dramatic impact on morbidity and mortality from diarrhea among children in high- and middle-income countries that have introduced the vaccine into their national immunization programs. Widespread introduction of rotavirus vaccine in developing countries is imminent and their full potential in reducing the global burden from severe childhood diarrhea may soon be realized. The objectives of this paper are to describe the remaining issues and challenges in ensuring the success of the global rotavirus vaccination program and to discuss further research needed to help address them.
Collapse
Affiliation(s)
- Jacqueline E Tate
- Centers for Disease Control and Prevention, 1600 Clifton Road, NE MS-A34, Atlanta, GA 30333, USA.
| | | | | | | | | | | | | | | |
Collapse
|
336
|
Contopoulos-Ioannidis DG, Seto I, Hamm MP, Thomson D, Hartling L, Ioannidis JPA, Curtis S, Constantin E, Batmanabane G, Klassen T, Williams K. Empirical evaluation of age groups and age-subgroup analyses in pediatric randomized trials and pediatric meta-analyses. Pediatrics 2012; 129 Suppl 3:S161-84. [PMID: 22661763 DOI: 10.1542/peds.2012-0055j] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND An important step toward improvement of the conduct of pediatric clinical research is the standardization of the ages of children to be included in pediatric trials and the optimal age-subgroups to be analyzed. METHODS We set out to evaluate empirically the age ranges of children, and age-subgroup analyses thereof, reported in recent pediatric randomized clinical trials (RCTs) and meta-analyses. First, we screened 24 RCTs published in Pediatrics during the first 6 months of 2011; second, we screened 188 pediatric RCTs published in 2007 in the Cochrane Central Register of Controlled Trials; third, we screened 48 pediatric meta-analyses published in the Cochrane Database of Systematic Reviews in 2011. We extracted information on age ranges and age-subgroups considered and age-subgroup differences reported. RESULTS The age range of children in RCTs published in Pediatrics varied from 0.1 to 17.5 years (median age: 5; interquartile range: 1.8-10.2) and only 25% of those presented age-subgroup analyses. Large variability was also detected for age ranges in 188 RCTs from the Cochrane Central Register of Controlled Trials, and only 28 of those analyzed age-subgroups. Moreover, only 11 of 48 meta-analyses had age-subgroup analyses, and in 6 of those, only different studies were included. Furthermore, most of these observed differences were not beyond chance. CONCLUSIONS We observed large variability in the age ranges and age-subgroups of children included in recent pediatric trials and meta-analyses. Despite the limited available data, some age-subgroup differences were noted. The rationale for the selection of particular age-subgroups deserves further study.
Collapse
Affiliation(s)
- Despina G Contopoulos-Ioannidis
- Department of Pediatrics, Division of Infectious Diseases, Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, California 94305, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
337
|
Lappalainen S, Ylitalo S, Arola A, Halkosalo A, Räsänen S, Vesikari T. Simultaneous presence of human herpesvirus 6 and adenovirus infections in intestinal intussusception of young children. Acta Paediatr 2012; 101:663-70. [PMID: 22296119 DOI: 10.1111/j.1651-2227.2012.02616.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIM This prospective study investigated the role of viral infections in the pathogenesis of intussusception, including human herpesvirus 6 (HHV-6), a known lymphotropic virus. METHODS Fifty-three children with intussusception treated in hospital were enroled, and children of comparable age and gender served as controls. Blood, stool and throat swab specimens, as well as mesenteric lymph nodes and pieces of intestine from patients requiring surgery were tested for various viruses by PCR methods. RESULTS Altogether, 85% of intussusception cases showed evidence of a recent or ongoing viral infection. Among the 53 intussusception cases, adenovirus was detected in 25 (47%), HHV-6 in 24 (45%), rhinovirus in 12 (23%), cytomegalovirus in 7 (13%), enterovirus in 4 (8%) and rotavirus in 3 (6%) patients. Of the 50 whole blood samples, 44% were positive for HHV-6 and of the 16 resected mesenteric lymph nodes, 50% were positive for HHV-6. Simultaneous presence of HHV-6 and adenovirus infection correlated significantly with intussusception (OR 12.1, 95% CI 2.2 to 66.5). CONCLUSIONS A statistically significant association was observed between adenovirus and childhood intussusception. HHV-6 was a common finding and occurred concomitantly with other viruses. A simultaneous infection with HHV-6 and adenovirus carried the highest risk for intussusception.
Collapse
Affiliation(s)
- Suvi Lappalainen
- Vaccine Research Center, University of Tampere Medical School, Tampere, Finland
| | | | | | | | | | | |
Collapse
|
338
|
Phua KB, Lim FS, Lau YL, Nelson EAS, Huang LM, Quak SH, Lee BW, Doorn LJV, Teoh YL, Tang H, Suryakiran P, Smolenov IV, Bock HL, Han HH. Rotavirus vaccine RIX4414 efficacy sustained during the third year of life: A randomized clinical trial in an Asian population. Vaccine 2012; 30:4552-7. [DOI: 10.1016/j.vaccine.2012.03.030] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 02/08/2012] [Accepted: 03/16/2012] [Indexed: 11/28/2022]
|
339
|
Diez Domingo J, Patrzalek M, Cantarutti L, Arnould B, Meunier J, Soriano-Gabarro M, Meyer N, Pirçon JY, Holl K. The impact of childhood acute rotavirus gastroenteritis on the parents' quality of life: prospective observational study in European primary care medical practices. BMC Pediatr 2012; 12:58. [PMID: 22650611 PMCID: PMC3495402 DOI: 10.1186/1471-2431-12-58] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 05/15/2012] [Indexed: 11/20/2022] Open
Abstract
Background Rotavirus (RV) is the commonest cause of acute gastroenteritis in infants and young children worldwide. A Quality of Life study was conducted in primary care in three European countries as part of a larger epidemiological study (SPRIK) to investigate the impact of paediatric rotavirus gastroenteritis (RVGE) on affected children and their parents. Methods A self-administered questionnaire was linguistically validated in Spanish, Italian and Polish. The questionnaire was included in an observational multicentre prospective study of 302 children aged <5 years presenting to a general practitioner or paediatrician for RVGE at centres in Spain, Italy or Poland. RV infection was confirmed by polymerase chain reaction (PCR) testing (n = 264). The questionnaire was validated and used to assess the emotional impact of paediatric RVGE on the parents. Results Questionnaire responses showed that acute RVGE in a child adversely affects the parents’ daily life as well as the child. Parents of children with RVGE experience worry, distress and impact on their daily activities. RVGE of greater clinical severity (assessed by the Vesikari scale) was associated with higher parental worries due to symptoms and greater changes in the child’s behaviour, and a trend to higher impact on parents’ daily activities and higher parental distress, together with a higher score on the symptom severity scale of the questionnaire. Conclusions Parents of a child with acute RVGE presenting to primary care experience worry, distress and disruptions to daily life as a result of the child’s illness. Prevention of this disease through prophylactic vaccination will improve the daily lives of parents and children.
Collapse
Affiliation(s)
- Javier Diez Domingo
- Centro Superior de Investigacion en Salud Publica (CSISP), Area de Investigación en Vacunas, Avda Catalunya 21, Valencia 46020, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
340
|
Holmgren J, Svennerholm AM. Vaccines against mucosal infections. Curr Opin Immunol 2012; 24:343-53. [PMID: 22580196 DOI: 10.1016/j.coi.2012.03.014] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 03/27/2012] [Indexed: 01/05/2023]
Abstract
There remains a great need to develop vaccines against many of the pathogens that infect mucosal tissues or have a mucosal port of entry. Parenteral vaccination may protect in some instances, but usually a mucosal vaccination route is necessary. Mucosal vaccines also have logistic advantages over injectable vaccines by being easier to administer, having less risk of transmitting infections and potentially being easier to manufacture. Still, however, only relatively few vaccines for human use are available: oral vaccines against cholera, typhoid, polio, and rotavirus, and a nasal vaccine against influenza. For polio, typhoid and influenza, in which the pathogens reach the blood stream, there is also an injectable vaccine alternative. A problem with available oral live vaccines is their reduced immunogenicity when used in developing countries; for instance, the efficacy of rotavirus vaccines correlates closely with the national per capita income. Research is needed to define the impact of factors such as malnutrition, aberrant intestinal microflora, concomitant infections, and preexisting immunity as well as of host genetic factors on the immunogenicity of these vaccines.
Collapse
Affiliation(s)
- Jan Holmgren
- University of Gothenburg Vaccine Research Institute (GUVAX) & Department of Microbiology and Immunology, The Sahlgrenska Academy at University of Gothenburg, Sweden.
| | | |
Collapse
|
341
|
Efficacy and immunogenicity of live-attenuated human rotavirus vaccine in breast-fed and formula-fed European infants. Pediatr Infect Dis J 2012; 31:509-13. [PMID: 22228235 DOI: 10.1097/inf.0b013e3182489cac] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Rotavirus is the main cause of severe gastroenteritis and diarrhea in infants and young children less than 5 years of age. Potential impact of breast-feeding on the efficacy and immunogenicity of human rotavirus G1P[8] vaccine was examined in this exploratory analysis. METHODS Healthy infants (N = 3994) aged 6-14 weeks who received 2 doses of human rotavirus vaccine/placebo according to a 0-1 or 0-2 month schedule were followed for rotavirus gastroenteritis during 2 epidemic seasons. Rotavirus IgA seroconversion rate (anti-IgA antibody concentration ≥ 20 mIU/mL) and geometric mean concentrations were measured prevaccination and 1-2 months post-dose 2. Vaccine efficacy against any and severe rotavirus gastroenteritis was analyzed according to the infants being breast-fed or exclusively formula-fed at the time of vaccination. RESULTS Antirotavirus IgA seroconversion rate was 85.5% (95% confidence interval [CI]: 82.4-88.3) in breast-fed and 89.2% (95% CI: 84.2-93) in exclusively formula-fed infants; geometric mean concentrations in the respective groups were 185.8 U/mL (95% CI: 161.4-213.9) and 231.5 U/mL (95% CI: 185.9-288.2). Vaccine efficacy was equally high in breast-fed and exclusively formula-fed children in the first season but fell in breast-fed infants in the second rotavirus season. During the combined 2-year efficacy follow-up period, vaccine efficacy against any rotavirus gastroenteritis was 76.2% (95% CI: 68.7-82.1) and 89.8% (95% CI: 77.6-95.9) and against severe rotavirus gastroenteritis 88.4% (95% CI: 81.6-93) and 98.1% (95% CI: 88.2-100) in the breast-fed and exclusively formula-fed infants, respectively. CONCLUSIONS The difference in immunogenicity of human rotavirus vaccine in breast-fed and exclusively formula-fed infants was small. Vaccine efficacy was equally high in breast-fed and exclusively formula-fed children in the first season. Breast-feeding seemed to reduce slightly the efficacy in the second season.
Collapse
|
342
|
Safety, reactogenicity and immunogenicity of the human rotavirus vaccine in preterm European Infants: a randomized phase IIIb study. Pediatr Infect Dis J 2012; 31:487-93. [PMID: 22228231 DOI: 10.1097/inf.0b013e3182490a2c] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Rotavirus disease is more severe in preterm infants than in full-term infants. This study assessed the safety, reactogenicity and immunogenicity of a human rotavirus vaccine, RIX4414, in European preterm infants. METHODS A total of 1009 preterm infants were randomized (2:1, vaccine:placebo) and stratified into 2 groups: 20% of early (27-30 weeks, group 1) and 80% of late (31-36 weeks, group 2) gestational age preterm infants in each group. Two doses of RIX4414/placebo were administered to these preterm infants according to the recommended chronologic age for full-term infants with an interval of 30-83 days between doses. Serious adverse events were recorded throughout the study period. Solicited and unsolicited adverse events were recorded for 15 and 31 days post-each dose. Antirotavirus IgA concentrations (enzyme-linked immunosorbent assay cutoff = 20 U/mL) and geometric mean concentration were determined pre-dose 1 and 30-83 days post-dose 2 in a subset of 300 infants. This study is registered with ClinicalTrials.gov, number NCT00420745 (eTrack106481). RESULTS Serious adverse events were reported at a similar frequency in both groups (P = 0.266). Fifty-seven infants reported at least 1 serious adverse event (5.1% [3.5-7.0] in the RIX4414 group and 6.8% [4.3-10.0] in the placebo group). During the 15-day postvaccination follow-up period, diarrhea, vomiting and fever occurred at a similar frequency in both groups; fever could have been due to concomitant vaccines. Five cases (RIX4414 = 3, Placebo = 2) of rotavirus gastroenteritis were reported. The onset of rotavirus gastroenteritis in the RIX4414 group was 1-5 days after vaccination (vaccine strain identified in all cases) and in the placebo group it was 3-4 days after receiving placebo (wild-type rotavirus identified from both cases). Antirotavirus IgA seroconversion rates at 30-83 days post-dose 2 were 85.7% (79.0-90.9) in the RIX4414 group and 16.0% (8.8-25.9) in the placebo group. Geometric mean concentrations were 202.2 U/mL (153.1-267.1) in the RIX4414 group and <20 U/mL in the placebo group. Seroconversion rate in groups 1 and 2 in RIX4414 recipients were 75.9% (95% confidence interval [CI]: 56.5-89.7%) and 88.1% (95% CI: 80.9-93.4%), respectively; the geometric mean concentrations in the respective groups were 110.2 U/mL (95% CI: 56.1-216.5) and 234.8 U/mL (95% CI: 173.4-318.0; exploratory analysis). CONCLUSIONS Two doses of RIX4414 were immunogenic and well-tolerated in European preterm infants.
Collapse
|
343
|
Nakagomi T, Nakagomi O, Dove W, Doan YH, Witte D, Ngwira B, Todd S, Steele AD, Neuzil KM, Cunliffe NA. Molecular characterization of rotavirus strains detected during a clinical trial of a human rotavirus vaccine in Blantyre, Malawi. Vaccine 2012; 30 Suppl 1:A140-51. [PMID: 22520123 PMCID: PMC3982048 DOI: 10.1016/j.vaccine.2011.09.119] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 08/31/2011] [Accepted: 09/28/2011] [Indexed: 12/13/2022]
Abstract
The human, G1P[8] rotavirus vaccine (Rotarix™) significantly reduced severe rotavirus gastroenteritis episodes in a clinical trial in South Africa and Malawi, but vaccine efficacy was lower in Malawi (49.5%) than reported in South Africa (76.9%) and elsewhere. The aim of this study was to examine the molecular relationships of circulating wild-type rotaviruses detected during the clinical trial in Malawi to RIX4414 (the strain contained in Rotarix™) and to common human rotavirus strains. Of 88 rotavirus-positive, diarrhoeal stool specimens, 43 rotaviruses exhibited identifiable RNA migration patterns when examined by polyacrylamide gel electrophoresis. The genes encoding VP7, VP4, VP6 and NSP4 of 5 representative strains possessing genotypes G12P[6], G1P[8], G9P[8], and G8P[4] were sequenced. While their VP7 (G) and VP4 (P) genotype designations were confirmed, the VP6 (I) and NSP4 (E) genotypes were either I1E1 or I2E2, indicating that they were of human rotavirus origin. RNA-RNA hybridization using 21 culture-adapted strains showed that Malawian rotaviruses had a genomic RNA constellation common to either the Wa-like or the DS-1 like human rotaviruses. Overall, the Malawi strains appear similar in their genetic make-up to rotaviruses described in countries where vaccine efficacy is greater, suggesting that the lower efficacy in Malawi is unlikely to be explained by the diversity of circulating strains.
Collapse
Affiliation(s)
- Toyoko Nakagomi
- Division of Molecular Epidemiology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, and the Global Centre of Excellence, Nagasaki University, Nagasaki, Japan
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Osamu Nakagomi
- Division of Molecular Epidemiology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, and the Global Centre of Excellence, Nagasaki University, Nagasaki, Japan
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Winifred Dove
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Yen Hai Doan
- Division of Molecular Epidemiology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, and the Global Centre of Excellence, Nagasaki University, Nagasaki, Japan
| | - Desiree Witte
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Bagrey Ngwira
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Stacy Todd
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | | | | | - Nigel A Cunliffe
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
344
|
Cunliffe NA, Witte D, Ngwira BM, Todd S, Bostock NJ, Turner AM, Chimpeni P, Victor JC, Steele AD, Bouckenooghe A, Neuzil KM. Efficacy of human rotavirus vaccine against severe gastroenteritis in Malawian children in the first two years of life: a randomized, double-blind, placebo controlled trial. Vaccine 2012; 30 Suppl 1:A36-43. [PMID: 22520135 PMCID: PMC3982044 DOI: 10.1016/j.vaccine.2011.09.120] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2011] [Revised: 09/20/2011] [Accepted: 09/28/2011] [Indexed: 10/28/2022]
Abstract
Rotavirus gastroenteritis is a major cause of morbidity and mortality among African infants and young children. A phase III, placebo-controlled, multi-centre clinical trial of a live, oral G1P[8] human rotavirus vaccine (RIX4414) undertaken in Malawi and South Africa significantly reduced the incidence of severe rotavirus gastroenteritis in the first year of life. We now report on vaccine efficacy in the Malawi cohort of children who were followed into the second year of life. A total of 1773 healthy infants were enrolled in Blantyre, Malawi into three groups. Two groups received three doses of RIX4414 or placebo at age 6, 10, and 14 weeks and the third group received placebo at 6 weeks and RIX4414 at age 10 and 14 weeks. Subjects were followed by weekly home visits for episodes of gastroenteritis until 1 year of age, and were then re-consented for further follow-up to 18-24 months of age. Severity of gastroenteritis episodes was graded according to the Vesikari scoring system. Seroconversion for anti-rotavirus IgA was determined on a subset of children by using ELISA on pre- and post-vaccine blood samples. Rotavirus VP7 (G) and VP4 (P) genotypes were determined by RT-PCR. A total of 70/1030 (6.8%, 95% CI 5.3-8.5) subjects in the pooled (2 dose plus 3 dose) RIX4414 group compared with 53/483 (11.0%, 8.3-14.1) subjects in the placebo group developed severe rotavirus gastroenteritis in the entire follow-up period (vaccine efficacy 38.1% (9.8-57.3)). The point estimate of efficacy in the second year of life (17.6%; -59.2 to 56.0) was lower than in the first year of life (49.4%; 19.2-68.3). There were non-significant trends towards a higher efficacy in the second year of life among children who received the three-dose schedule compared with the two-dose schedule, and a higher anti-rotavirus IgA seroresponse rate in the three-dose RIX4414 group. Rotavirus strains detected included genotype G12 (31%); G9 (23%); and G8 (18%); only 18% of strains belonged to the G1P[8] genotype. While the optimal dosing schedule of RIX4414 in African infants requires further investigation, vaccination with RIX4414 significantly reduced the incidence of severe gastroenteritis caused by diverse rotavirus strains in an impoverished African population with high rotavirus disease burden in the first two years of life.
Collapse
Affiliation(s)
- Nigel A Cunliffe
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Daulby Street, Liverpool L69 3GA, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
345
|
Chen SC, Tan LB, Huang LM, Chen KT. Rotavirus infection and the current status of rotavirus vaccines. J Formos Med Assoc 2012; 111:183-93. [DOI: 10.1016/j.jfma.2011.09.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 09/16/2011] [Accepted: 09/28/2011] [Indexed: 12/12/2022] Open
|
346
|
Influence of oral polio vaccines on performance of the monovalent and pentavalent rotavirus vaccines. Vaccine 2012; 30 Suppl 1:A30-5. [DOI: 10.1016/j.vaccine.2011.11.093] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 10/28/2011] [Accepted: 11/23/2011] [Indexed: 11/18/2022]
|
347
|
Efficacy and immunogenicity of two or three dose rotavirus-vaccine regimen in South African children over two consecutive rotavirus-seasons: A randomized, double-blind, placebo-controlled trial. Vaccine 2012; 30 Suppl 1:A44-51. [DOI: 10.1016/j.vaccine.2011.08.080] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Revised: 08/08/2011] [Accepted: 08/15/2011] [Indexed: 11/21/2022]
|
348
|
Verma R, Khanna P, Chawla S. Rotavirus vaccine can save millions of children's lives in developing countries. Hum Vaccin Immunother 2012; 8:272-4. [PMID: 22426375 DOI: 10.4161/hv.18390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Ramesh Verma
- Department of Community Medicine, Pt. B.D. Sharma PGIMS, Rohtak, Haryana India.
| | | | | |
Collapse
|
349
|
Arientova S, Schramlova J, Ambrozova H, Maresova V, Holub M. Electron microscopy in the diagnosis of viral gastroenteritis in hospitalised children in the Czech Republic. Folia Microbiol (Praha) 2012; 57:177-82. [PMID: 22430885 PMCID: PMC7090978 DOI: 10.1007/s12223-012-0109-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 03/02/2012] [Indexed: 12/03/2022]
Abstract
Our study has been aimed at demonstrating the main role of viruses in the aetiology of acute gastroenteritis in children less than 5 years old and at pointing out the diagnostic potential of electron microscopy in the diagnosis of viral gastroenteritis. A prospective study was conducted to analyse the aetiology of diarrhoeal diseases in children less than 5 years of age admitted to the Department of Infectious Diseases between September 2006 and December 2008. All children were tested by faecal culture, latex agglutination and electron microscopy. A total of 832 children were included in the study. An aetiological agent was detected in 788 children (94.6 %). A bacterial aetiology was found in 22 (2.6 %) children and bacterial–viral co-infection was found in 146 (17.6 %) patients. The most frequent causative agents of gastroenteritis in children were viruses, which were detected in 620 (74.5 %) patients. The main causes of viral gastroenteritis were rotaviruses (detected in 410 children), followed by caliciviruses (42), coronaviruses (28), adenoviruses (19) and astroviruses (14). Dual viral infections were detected in 107 children, with rotavirus–calicivirus co-infection being the most common. Electron microscopy proved to be a more sensitive method in comparison with the latex agglutination test for the diagnosis of rotaviruses and adenoviruses. The major role of viruses in diarrhoeal diseases among children under 5 years of age in the Czech Republic has been confirmed. The diagnostic potential of electron microscopy, particularly in small outbreaks of gastroenteritis, was clearly shown.
Collapse
Affiliation(s)
- Simona Arientova
- Department of Infectious and Tropical Diseases, Charles University in Prague, First Faculty of Medicine, University Hospital Bulovka, Prague, Czech Republic.
| | | | | | | | | |
Collapse
|
350
|
O'Ryan M, Lucero Y, Linhares AC. Rotarix®: vaccine performance 6 years postlicensure. Expert Rev Vaccines 2012; 10:1645-59. [PMID: 22085167 DOI: 10.1586/erv.11.152] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Rotarix(®) was first licensed in 2004 and rapidly introduced into private and public markets worldwide. In a previous 2009 article, we reviewed the impact of rotavirus-associated disease, the rationale for different vaccines, prelicensure efficacy studies and cost-effectiveness studies for Rotarix. As of September 2011, Rotarix had been licensed in 123 countries in the Americas, Europe, Australia, Africa and Asia, of which 27 have incorporated the vaccine into national or regional immunization programs. The current review intends to provide the reader with further insight into this vaccine, focusing mainly on the new information obtained after a 6-year postlicensure period. This review will provide only a brief summary of prelicensure studies extensively discussed in the previous publication and refer, in more depth, to the worldwide experience with the vaccine, vaccine impact, and safety observed in effectiveness and observational studies, including a particular analysis on protection against rotavirus G2P[4].
Collapse
Affiliation(s)
- Miguel O'Ryan
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | | | | |
Collapse
|