101
|
Soares‐Weiser K, Bergman H, Henschke N, Pitan F, Cunliffe N. Vaccines for preventing rotavirus diarrhoea: vaccines in use. Cochrane Database Syst Rev 2019; 2019:CD008521. [PMID: 31684685 PMCID: PMC6816010 DOI: 10.1002/14651858.cd008521.pub5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Rotavirus results in more diarrhoea-related deaths in children under five years 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. Rotavirus vaccines that have been prequalified by the World Health Organization (WHO) include a monovalent vaccine (RV1; Rotarix, GlaxoSmithKline), a pentavalent vaccine (RV5; RotaTeq, Merck), and, more recently, another monovalent vaccine (Rotavac, Bharat Biotech). OBJECTIVES To evaluate rotavirus vaccines prequalified by the WHO (RV1, RV5, and Rotavac) for their efficacy and safety in children. SEARCH METHODS On 4 April 2018 we searched MEDLINE (via PubMed), the Cochrane Infectious Diseases Group Specialized Register, CENTRAL (published in the Cochrane Library), Embase, LILACS, and BIOSIS. We also searched the WHO ICTRP, ClinicalTrials.gov, clinical trial reports from manufacturers' websites, and reference lists of included studies and relevant systematic reviews. SELECTION CRITERIA We selected randomized controlled trials (RCTs) in children comparing rotavirus vaccines prequalified for use by the WHO versus placebo or no intervention. DATA COLLECTION AND ANALYSIS Two review authors independently assessed trial eligibility and assessed risks of bias. One review author extracted data and a second author cross-checked them. We combined dichotomous data using the risk ratio (RR) and 95% confidence interval (CI). We stratified the analysis by country mortality rate and used GRADE to evaluate evidence certainty. MAIN RESULTS Fifty-five trials met the inclusion criteria and enrolled a total of 216,480 participants. Thirty-six trials (119,114 participants) assessed RV1, 15 trials (88,934 participants) RV5, and four trials (8432 participants) Rotavac. RV1 Children vaccinated and followed up the first year of life In low-mortality countries, RV1 prevents 84% of severe rotavirus diarrhoea cases (RR 0.16, 95% CI 0.09 to 0.26; 43,779 participants, 7 trials; high-certainty evidence), and probably prevents 41% of cases of severe all-cause diarrhoea (RR 0.59, 95% CI 0.47 to 0.74; 28,051 participants, 3 trials; moderate-certainty evidence). In high-mortality countries, RV1 prevents 63% of severe rotavirus diarrhoea cases (RR 0.37, 95% CI 0.23 to 0.60; 6114 participants, 3 trials; high-certainty evidence), and 27% of severe all-cause diarrhoea cases (RR 0.73, 95% CI 0.56 to 0.95; 5639 participants, 2 trials; high-certainty evidence). Children vaccinated and followed up for two years In low-mortality countries, RV1 prevents 82% of severe rotavirus diarrhoea cases (RR 0.18, 95% CI 0.14 to 0.23; 36,002 participants, 9 trials; high-certainty evidence), and probably prevents 37% of severe all-cause diarrhoea episodes (rate ratio 0.63, 95% CI 0.56 to 0.71; 39,091 participants, 2 trials; moderate-certainty evidence). In high-mortality countries RV1 probably prevents 35% of severe rotavirus diarrhoea cases (RR 0.65, 95% CI 0.51 to 0.83; 13,768 participants, 2 trials; high-certainty evidence), and 17% of severe all-cause diarrhoea cases (RR 0.83, 95% CI 0.72 to 0.96; 2764 participants, 1 trial; moderate-certainty evidence). No increased risk of serious adverse events (SAE) was detected (RR 0.88 95% CI 0.83 to 0.93; high-certainty evidence). There were 30 cases of intussusception reported in 53,032 children after RV1 vaccination and 28 cases in 44,214 children after placebo or no intervention (RR 0.70, 95% CI 0.46 to 1.05; low-certainty evidence). RV5 Children vaccinated and followed up the first year of life In low-mortality countries, RV5 probably prevents 92% of severe rotavirus diarrhoea cases (RR 0.08, 95% CI 0.03 to 0.22; 4132 participants, 5 trials; moderate-certainty evidence). We did not identify studies reporting on severe all-cause diarrhoea in low-mortality countries. In high-mortality countries, RV5 prevents 57% of severe rotavirus diarrhoea (RR 0.43, 95% CI 0.29 to 0.62; 5916 participants, 2 trials; high-certainty evidence), but there is probably little or no difference between vaccine and placebo for severe all-cause diarrhoea (RR 0.80, 95% CI 0.58 to 1.11; 1 trial, 4085 participants; moderate-certainty evidence). Children vaccinated and followed up for two years In low-mortality countries, RV5 prevents 82% of severe rotavirus diarrhoea cases (RR 0.18, 95% CI 0.08 to 0.39; 7318 participants, 4 trials; moderate-certainty evidence). We did not identify studies reporting on severe all-cause diarrhoea in low-mortality countries. In high-mortality countries, RV5 prevents 41% of severe rotavirus diarrhoea cases (RR 0.59, 95% CI 0.43 to 0.82; 5885 participants, 2 trials; high-certainty evidence), and 15% of severe all-cause diarrhoea cases (RR 0.85, 95% CI 0.75 to 0.98; 5977 participants, 2 trials; high-certainty evidence). No increased risk of serious adverse events (SAE) was detected (RR 0.93 95% CI 0.86 to 1.01; moderate to high-certainty evidence). There were 16 cases of intussusception in 43,629 children after RV5 vaccination and 20 cases in 41,866 children after placebo (RR 0.77, 95% CI 0.41 to 1.45; low-certainty evidence). Rotavac Children vaccinated and followed up the first year of life Rotavac has not been assessed in any RCT in countries with low child mortality. In India, a high-mortality country, Rotavac probably prevents 57% of severe rotavirus diarrhoea cases (RR 0.43, 95% CI 0.30 to 0.60; 6799 participants, moderate-certainty evidence); the trial did not report on severe all-cause diarrhoea at one-year follow-up. Children vaccinated and followed up for two years Rotavac probably prevents 54% of severe rotavirus diarrhoea cases in India (RR 0.46, 95% CI 0.35 to 0.60; 6541 participants, 1 trial; moderate-certainty evidence), and 16% of severe all-cause diarrhoea cases (RR 0.84, 95% CI 0.71 to 0.98; 6799 participants, 1 trial; moderate-certainty evidence). No increased risk of serious adverse events (SAE) was detected (RR 0.93 95% CI 0.85 to 1.02; moderate-certainty evidence). There were eight cases of intussusception in 5764 children after Rotavac vaccination and three cases in 2818 children after placebo (RR 1.33, 95% CI 0.35 to 5.02; very low-certainty evidence). There was insufficient evidence of an effect on mortality from any rotavirus vaccine (198,381 participants, 44 trials; low- to very low-certainty evidence), as the trials were not powered to detect an effect at this endpoint. AUTHORS' CONCLUSIONS RV1, RV5, and Rotavac prevent episodes of rotavirus diarrhoea. Whilst the relative effect estimate is smaller in high-mortality than in low-mortality countries, there is a greater number of episodes prevented in these settings as the baseline risk is much higher. We found no increased risk of serious adverse events. 21 October 2019 Up to date All studies incorporated from most recent search All published trials found in the last search (4 Apr, 2018) were included and 15 ongoing studies are currently awaiting completion (see 'Characteristics of ongoing studies').
Collapse
Affiliation(s)
- Karla Soares‐Weiser
- CochraneEditorial & Methods DepartmentSt Albans House, 57 ‐ 59 HaymarketLondonUKSW1Y 4QX
| | - Hanna Bergman
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
| | - Nicholas Henschke
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
| | - Femi Pitan
- Chevron Corporation2 Chevron DriveLekkiLagosNigeria
| | - Nigel Cunliffe
- University of LiverpoolInstitute of Infection and Global Health, Faculty of Health and Life SciencesLiverpoolUKL69 7BE
| | | |
Collapse
|
102
|
Lu HL, Ding Y, Goyal H, Xu HG. Association Between Rotavirus Vaccination and Risk of Intussusception Among Neonates and Infants: A Systematic Review and Meta-analysis. JAMA Netw Open 2019; 2:e1912458. [PMID: 31584679 PMCID: PMC6784808 DOI: 10.1001/jamanetworkopen.2019.12458] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 08/14/2019] [Indexed: 12/19/2022] Open
Abstract
IMPORTANCE The conclusions from the multiple randomized clinical trials exploring the relationship between development of intussusception and rotavirus vaccination among neonates and infants have been controversial. OBJECTIVE To evaluate the association between rotavirus vaccination and risk of intussusception. DATA SOURCES For this systematic review and meta-analysis, PubMed, Web of Science, Cochrane library, and Embase databases were searched from January 1, 1999, through December 31, 2018, using no language restrictions. The search terms were rotavirus or RV (rotavirus vaccine) or HRV (human rotavirus vaccine), vaccin*, and intussusception. STUDY SELECTION Randomized clinical trials of neonates and infants that compared the risk of intussusception after the vaccination with a placebo group were included. DATA EXTRACTION AND SYNTHESIS A fixed-effects model was used to pool the data. Statistical heterogeneity was assessed with Q test and I2 statistic; relative risk (RR), risk difference (RD), and 95% CIs were calculated using the Mantel-Haenszel method. MAIN OUTCOMES AND MEASURES The main outcome was the diagnosis of intussusception in the analysis. The pooled and subtotal results of RR, RD, and 95% CI for the risk of intussusception were estimated at 31 days, 1 year, and 2 years after vaccination. RESULTS A total of 25 randomized clinical trials including 200 594 participants (104 647 receiving vaccine and 95 947 receiving placebo) in 33 countries from 4 continents were included in this meta-analysis. Twenty cases of definite intussusception were diagnosed within 31 days after rotavirus vaccination, with 11 cases (55%) in the vaccine group and 9 cases (45%) in the placebo group (RD, 0.17 per 10 000 infants [95% CI, -1.16 to 1.50 per 10 000 infants], P = .80; RR, 1.14 [95% CI, 0.49 to 2.64], P = .77). Seventy-four cases were reported within 1 year, with 37 cases (50%) in the vaccine group and 37 cases (50%) in the placebo group (RD, -0.65 per 10 000 infants [95% CI, -2.68 to 1.39 per 10 000 infants], P = .53; RR, 0.84 [95% CI, 0.53 to 1.32], P = .45). Fifty-nine cases were reported within 2 years, with 29 cases (49%) in the vaccine group and 30 cases (51%) in the placebo group (RD, -0.48 per 10 000 infants [95% CI, -3.64 to 2.69 per 10 000 infants], P = .77; RR, 0.91 [95% CI, 0.55 to 1.52], P = .73). CONCLUSIONS AND RELEVANCE Results of this systematic review and meta-analysis suggest that monovalent, pentavalent, monovalent human-bovine, oral bovine pentavalent, and human neonatal rotavirus vaccination was not associated with an elevated risk of intussusception among neonates or infants.
Collapse
Affiliation(s)
- Hai-Ling Lu
- Department of Laboratory Medicine, Yancheng Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, China
| | - Ying Ding
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hemant Goyal
- Department of Internal Medicine, Mercer University School of Medicine, Macon, Georgia
| | - Hua-Guo Xu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
103
|
Payne DC, Englund JA, Weinberg GA, Halasa NB, Boom JA, Staat MA, Selvarangan R, Azimi PH, Klein EJ, Szilagyi PG, Chappell J, Sahni LC, McNeal M, Harrison CJ, Moffatt ME, Johnston SH, Mijatovic-Rustempasic S, Esona MD, Tate JE, Curns AT, Wikswo ME, Sulemana I, Bowen MD, Parashar UD. Association of Rotavirus Vaccination With Inpatient and Emergency Department Visits Among Children Seeking Care for Acute Gastroenteritis, 2010-2016. JAMA Netw Open 2019; 2:e1912242. [PMID: 31560386 PMCID: PMC6777243 DOI: 10.1001/jamanetworkopen.2019.12242] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
IMPORTANCE Rotavirus vaccines have been recommended for universal US infant immunization for more than 10 years, and understanding their effectiveness is key to the continued success of the US rotavirus vaccine immunization program. OBJECTIVE To assess the association of RotaTeq (RV5) and Rotarix (RV1) with inpatient and emergency department (ED) visits for rotavirus infection. DESIGN, SETTING, AND PARTICIPANTS This case-control vaccine effectiveness study was performed at inpatient and ED clinical settings in 7 US pediatric medical institutions from November 1, 2009, through June 30, 2016. Children younger than 5 years seeking medical care for acute gastroenteritis were enrolled. Clinical and epidemiologic data, vaccination verification, and results of stool sample tests for laboratory-confirmed rotavirus were collected. Data were analyzed from November 1, 2009, through June 30, 2016. MAIN OUTCOMES AND MEASURES Rotavirus vaccine effectiveness for preventing rotavirus-associated inpatient and ED visits over time for each licensed vaccine, stratified by clinical severity and age. RESULTS Among the 10 813 children included (5927 boys [54.8%] and 4886 girls [45.2%]; median [range] age, 21 [8-59] months), RV5 and RV1 analyses found that compared with controls, rotavirus-positive cases were more often white (RV5, 535 [62.2%] vs 3310 [57.7%]; RV1, 163 [43.1%] vs 864 [35.1%]), privately insured (RV5, 620 [72.1%] vs 4388 [76.5%]; RV1, 305 [80.7%] vs 2140 [87.0%]), and older (median [range] age for RV5, 26 [8-59] months vs 21 [8-59] months; median [range] age for RV1, 22 [8-59] months vs 19 [8-59] months) but did not differ by sex. Among 1193 rotavirus-positive cases and 9620 rotavirus-negative controls, at least 1 dose of any rotavirus vaccine was 82% (95% CI, 77%-86%) protective against rotavirus-associated inpatient visits and 75% (95% CI, 71%-79%) protective against rotavirus-associated ED visits. No statistically significant difference during this 7-year period was observed for either rotavirus vaccine. Vaccine effectiveness against inpatient and ED visits was 81% (95% CI, 78%-84%) for RV5 (3 doses) and 78% (95% CI, 72%-82%) for RV1 (2 doses) among the study population. A mixed course of both vaccines provided 86% (95% CI, 74%-93%) protection. Rotavirus patients who were not vaccinated had severe infections 4 times more often than those who were vaccinated (74 of 426 [17.4%] vs 28 of 605 [4.6%]; P < .001), and any dose of rotavirus vaccine was 65% (95% CI, 56%-73%) effective against mild infections, 81% (95% CI, 76%-84%) against moderate infections, and 91% (95% CI, 85%-95%) against severe infections. CONCLUSIONS AND RELEVANCE Evidence from this large postlicensure study of rotavirus vaccine performance in the United States from 2010 to 2016 suggests that RV5 and RV1 rotavirus vaccines continue to perform well, particularly in preventing inpatient visits and severe infections and among younger children.
Collapse
Affiliation(s)
- Daniel C. Payne
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Janet A. Englund
- Department of Pediatrics, Seattle Children’s Hospital, Seattle, Washington
- Seattle Children’s Research Institute, Seattle Children’s Hospital, Seattle, Washington
| | - Geoffrey A. Weinberg
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Natasha B. Halasa
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Julie A. Boom
- Immunization Project, Texas Children’s Hospital, Houston
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Mary Allen Staat
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Rangaraj Selvarangan
- Department of Pathology and Laboratory Medicine, Children’s Mercy Hospitals and Clinics, Children’s Mercy, Kansas City, Missouri
| | - Parvin H. Azimi
- Department of Infectious Disease, UCSF (University of California, San Francisco) Benioff Children’s Hospital Oakland, Oakland
| | - Eileen J. Klein
- Department of Pediatrics, Seattle Children’s Hospital, Seattle, Washington
- Seattle Children’s Research Institute, Seattle Children’s Hospital, Seattle, Washington
| | - Peter G. Szilagyi
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York
- Department of Pediatrics, UCLA (University of California, Los Angeles)
| | - James Chappell
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Leila C. Sahni
- Immunization Project, Texas Children’s Hospital, Houston
| | - Monica McNeal
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Christopher J. Harrison
- Department of Infectious Disease, UCSF (University of California, San Francisco) Benioff Children’s Hospital Oakland, Oakland
| | - Mary E. Moffatt
- Division of Infectious Diseases, Children’s Mercy, Kansas City, Missouri
| | | | - Slavica Mijatovic-Rustempasic
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Mathew D. Esona
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jacqueline E. Tate
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Aaron T. Curns
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Mary E. Wikswo
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Iddrisu Sulemana
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Michael D. Bowen
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Umesh D. Parashar
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
104
|
Boix-Amorós A, Collado MC, Van't Land B, Calvert A, Le Doare K, Garssen J, Hanna H, Khaleva E, Peroni DG, Geddes DT, Kozyrskyj AL, Warner JO, Munblit D. Reviewing the evidence on breast milk composition and immunological outcomes. Nutr Rev 2019; 77:541-556. [PMID: 31111150 DOI: 10.1093/nutrit/nuz019] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A large number of biologically active components have been found in human milk (HM), and in both human and animal models, studies have provided some evidence suggesting that HM composition can be altered by maternal exposures, subsequently influencing health outcomes for the breastfed child. Evidence varies from the research studies on whether breastfeeding protects the offspring from noncommunicable diseases, including those associated with immunological dysfunction. It has been hypothesized that the conflicting evidence results from HM composition variations, which contain many immune active molecules, oligosaccharides, lactoferrin, and lysozyme in differing concentrations, along with a diverse microbiome. Determining the components that influence infant health outcomes in terms of both short- and long-term sequelae is complicated by a lack of understanding of the environmental factors that modify HM constituents and thereby offspring outcomes. Variations in HM immune and microbial composition (and the differing infantile responses) may in part explain the controversies that are evidenced in studies that aim to evaluate the prevalence of allergy by prolonged and exclusive breastfeeding. HM is a "mixture" of immune active factors, oligosaccharides, and microbes, which all may influence early immunological outcomes. This comprehensive review provides an in-depth overview of existing evidence on the studied relationships between maternal exposures, HM composition, vaccine responses, and immunological outcomes.
Collapse
Affiliation(s)
- Alba Boix-Amorós
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
| | - Belinda Van't Land
- Department of Immunology, Danone Nutricia Research, Utrecht, the Netherlands
- Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Anna Calvert
- Paediatric Infectious Diseases Research Group, St George's University of London, London, United Kingdom
| | - Kirsty Le Doare
- Paediatric Infectious Diseases Research Group, St George's University of London, London, United Kingdom
- Imperial College London, London, United Kingdom
- Public Health England, Porton Down, United Kingdom, and the MRC Unit, Fajara, Gambia
| | - Johan Garssen
- Department of Immunology, Danone Nutricia Research, Utrecht, the Netherlands
- Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | | | - Ekaterina Khaleva
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- University of Southampton, Southampton, UK
| | - Diego G Peroni
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Department of Clinical and Experimental Medicine, Section of Paediatrics, University of Pisa, Pisa, Italy
| | - Donna T Geddes
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- School of Molecular Sciences, the University of Western Australia, Perth, Australia
| | - Anita L Kozyrskyj
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Department of Pediatrics, Department of Obstetrics & Gynecology, Faculty of Medicine & Dentistry, School of Public Health, University of Alberta, Alberta, Canada
| | - John O Warner
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Imperial College London, London, United Kingdom
- National Institute for Health Research, Collaboration for Leadership in Applied Health Research and Care for NW London, London, United Kingdom
| | - Daniel Munblit
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Imperial College London, London, United Kingdom
- Department of Pediatrics, Sechenov University, Moscow, Russia, and the Solov'ev Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| |
Collapse
|
105
|
Rogawski ET, Platts-Mills JA, Colgate ER, Haque R, Zaman K, Petri WA, Kirkpatrick BD. Quantifying the Impact of Natural Immunity on Rotavirus Vaccine Efficacy Estimates: A Clinical Trial in Dhaka, Bangladesh (PROVIDE) and a Simulation Study. J Infect Dis 2019. [PMID: 29514306 DOI: 10.1093/infdis/jix668] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Background The low efficacy of rotavirus vaccines in clinical trials performed in low-resource settings may be partially explained by acquired immunity from natural exposure, especially in settings with high disease incidence. Methods In a clinical trial of monovalent rotavirus vaccine in Bangladesh, we compared the original per-protocol efficacy estimate to efficacy derived from a recurrent events survival model in which children were considered naturally exposed and potentially immune after their first rotavirus diarrhea (RVD) episode. We then simulated trial cohorts to estimate the expected impact of prior exposure on efficacy estimates for varying rotavirus incidence rates and vaccine efficacies. Results Accounting for natural immunity increased the per-protocol vaccine efficacy estimate against severe RVD from 63.1% (95% confidence interval [CI], 33.0%-79.7%) to 70.2% (95% CI, 44.5%-84.0%) in the postvaccination period, and original year 2 efficacy was underestimated by 14%. The simulations demonstrated that this expected impact increases linearly with RVD incidence, will be greatest for vaccine efficacies near 50%, and can reach 20% in settings with high incidence and low efficacy. Conclusions High rotavirus incidence leads to predictably lower vaccine efficacy estimates due to the acquisition of natural immunity in unvaccinated children, and this phenomenon should be considered when comparing efficacy estimates across settings. Clinical Trials Registration NCT01375647.
Collapse
Affiliation(s)
- Elizabeth T Rogawski
- Department of Public Health Sciences, University of Virginia, Charlottesville.,Division of Infectious Diseases and International Health, University of Virginia, Charlottesville
| | - James A Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville
| | - E Ross Colgate
- Department of Medicine and Vaccine Testing Center, University of Vermont College of Medicine, Burlington
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - K Zaman
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - William A Petri
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville
| | - Beth D Kirkpatrick
- Department of Medicine and Vaccine Testing Center, University of Vermont College of Medicine, Burlington
| |
Collapse
|
106
|
New pentavalent rotavirus vaccine shows little efficacy against diarrhea. Vaccine 2019; 37:3945. [DOI: 10.1016/j.vaccine.2018.05.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 11/23/2022]
|
107
|
Martinez-Gutierrez M, Arcila-Quiceno V, Trejos-Suarez J, Ruiz-Saenz J. Prevalence and molecular typing of rotavirus in children with acute diarrhoea in Northeastern Colombia. Rev Inst Med Trop Sao Paulo 2019; 61:e34. [PMID: 31269110 PMCID: PMC6609135 DOI: 10.1590/s1678-9946201961034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/26/2019] [Indexed: 12/15/2022] Open
Abstract
After the introduction of the rotavirus vaccine, the number of
rotavirus-associated deaths and the predicted annual rotavirus detection rate
had slightly declined worldwide. Taking in account that in Colombia, Rotarix
vaccine was introduced in 2009, the purpose of this study was to evaluate the
presence of rotavirus A in children under five years who were treated for acute
diarrhoea in Bucaramanga, Colombia and, moreover, to determine the genotypes of
rotavirus present in those children. We performed an analytical cross-sectional
study of rotavirus A in faecal samples from children up to five years of age.
Stool samples were screened for rotavirus A using a lateral-flow
immunochromatographic assay and confirmed using a VP6 sandwich ELISA. Genotyping
of rotavirus A-positive samples was performed by PCR and sequencing of VP7 and
VP4 genes. The overall prevalence of rotavirus was 30.53% (95% confidence
interval [CI] 21.2 - 39.7). Most of the children with rotavirus (86.2%) had
received two doses of the rotavirus vaccine. G3 strains accounted for the vast
majority of cases (82.8%), followed by G12 strains (13.8%) and G3/G9
coinfections (3.4%). Among the P genotypes, P[8] was the most prevalent (69%),
followed by P[9] (31%). The most common G[P] genotype combination was G3P[8],
followed by G3P[9]. The main finding in this study was that rotavirus, in a
Colombian region, is still an important pathogen in children under five years
old, previously vaccinated. The results showed that different factors, such as
kindergarten attendance, could explain the epidemiology and transmission of
rotavirus in Bucaramanga.
Collapse
Affiliation(s)
- Marlen Martinez-Gutierrez
- Universidad Cooperativa de Colombia, Grupo de Investigación en Ciencias Animales, Bucaramanga, Colombia
| | - Victor Arcila-Quiceno
- Universidad Cooperativa de Colombia, Grupo de Investigación en Ciencias Animales, Bucaramanga, Colombia
| | - Juanita Trejos-Suarez
- Universidad de Santander, Facultad de Ciencias de la Salud, Programa de Bacteriología y Laboratorio Clínico, Grupo de Investigación en Manejo Clínico, Bucaramanga, Colombia
| | - Julian Ruiz-Saenz
- Universidad Cooperativa de Colombia, Grupo de Investigación en Ciencias Animales, Bucaramanga, Colombia
| |
Collapse
|
108
|
Clark A, van Zandvoort K, Flasche S, Sanderson C, Bines J, Tate J, Parashar U, Jit M. Efficacy of live oral rotavirus vaccines by duration of follow-up: a meta-regression of randomised controlled trials. THE LANCET. INFECTIOUS DISEASES 2019; 19:717-727. [PMID: 31178289 PMCID: PMC6595176 DOI: 10.1016/s1473-3099(19)30126-4] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/07/2019] [Accepted: 02/15/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND The duration of protection offered by rotavirus vaccines varies across the world, and this variation is important to understanding and predicting the effects of the vaccines. There is now a large body of evidence on the efficacy of live oral rotavirus vaccines in different settings, but these data have never been synthesised to obtain robust estimates of efficacy by duration of follow-up. Our aim is to estimate the efficacy of live oral rotavirus vaccines at each point during follow-up and by mortality stratum. METHODS In our meta-regression study, we identified all randomised controlled trials of rotavirus vaccines published until April 4, 2018, using the results of a Cochrane systematic review, and cross checked these studies against those identified by another systematic review. We excluded trials that were based on special populations, trials without an infant schedule, and trials without clear reporting of numbers of enrolled infants and events in different periods of follow-up. For all reported periods of follow-up, we extracted the mean duration of follow-up (time since administration of the final dose of rotavirus vaccination), the number of enrolled infants, and case counts for rotavirus-positive severe gastroenteritis in both non-vaccinated and vaccinated groups. We used a Bayesian hierarchical Poisson meta-regression model to estimate the pooled cumulative vaccine efficacy (VE) and its waning with time for three mortality strata. We then converted these VE estimates into instantaneous VE (iVE). FINDINGS In settings with low mortality (15 observations), iVE pooled for infant schedules of Rotarix and RotaTeq was 98% (95% credibility interval 93-100) 2 weeks following the final dose of vaccination and 94% (87-98) after 12 months. In medium-mortality settings (11 observations), equivalent estimates were 82% (74-92) after 2 weeks and 77% (67-84) after 12 months. In settings with high mortality (24 observations), there were five different vaccines with observation points for infant schedules. The pooled iVE was 66% (48-81) after 2 weeks of follow-up and 44% (27-59) after 12 months. INTERPRETATION Rotavirus vaccine efficacy is lower and wanes more rapidly in high-mortality settings than in low-mortality settings, but the earlier peak age of disease in high-mortality settings means that live oral rotavirus vaccines are still likely to provide substantial benefit. FUNDING Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Andrew Clark
- London School of Hygiene and Tropical Medicine, London, UK.
| | | | - Stefan Flasche
- London School of Hygiene and Tropical Medicine, London, UK
| | | | - Julie Bines
- Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia; Department of Gastroenterology and Clinical Nutrition, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Jacqueline Tate
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Umesh Parashar
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Mark Jit
- London School of Hygiene and Tropical Medicine, London, UK; Modelling and Economics Unit, Public Health England, London, UK
| |
Collapse
|
109
|
Brewster R, Tamburini FB, Asiimwe E, Oduaran O, Hazelhurst S, Bhatt AS. Surveying Gut Microbiome Research in Africans: Toward Improved Diversity and Representation. Trends Microbiol 2019; 27:824-835. [PMID: 31178123 DOI: 10.1016/j.tim.2019.05.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/09/2019] [Accepted: 05/15/2019] [Indexed: 12/16/2022]
Abstract
Descriptive and translational investigations into the human gut microbiome (GM) are rapidly expanding; however, studies are largely restricted to industrialized populations in the USA and Europe. Little is known about microbial variability and its implications for health and disease in other parts of the world. Populations in Africa are particularly underrepresented. What limited research has been performed has focused on a few subject domains, including the impact of long-term lifestyle and dietary factors on GM ecology, its maturation during infancy, and the interrelationships between the microbiome, infectious disease, and undernutrition. Recently, international consortia have laid the groundwork for large-scale genomics and microbiome studies on the continent, with a particular interest in the epidemiologic transition to noncommunicable disease. Here, we survey the current landscape of GM scholarship in Africa and propose actionable recommendations to improve research capacity and output.
Collapse
Affiliation(s)
- Ryan Brewster
- School of Medicine, Stanford University, Stanford, CA, USA
| | | | - Edgar Asiimwe
- School of Medicine, Stanford University, Stanford, CA, USA
| | - Ovokeraye Oduaran
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
| | - Scott Hazelhurst
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa; School of Electrical and Information Engineering, University of the Witwatersrand, Johannesburg, South Africa.
| | - Ami S Bhatt
- School of Medicine, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA; Department of Medicine (Hematology), Stanford University, Stanford, CA, USA.
| |
Collapse
|
110
|
Rojas M, Dias HG, Gonçalves JLS, Manchego A, Rosadio R, Pezo D, Santos N. Genetic diversity and zoonotic potential of rotavirus A strains in the southern Andean highlands, Peru. Transbound Emerg Dis 2019; 66:1718-1726. [PMID: 31002476 DOI: 10.1111/tbed.13207] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/06/2019] [Accepted: 04/12/2019] [Indexed: 12/17/2022]
Abstract
Interspecies transmission is an important mechanism of evolution and contributes to rotavirus A (RVA) diversity. In order to evaluate the detection frequency, genetic diversity, epidemiological characteristics and zoonotic potential of RVA strains in faecal specimens from humans and animals cohabiting in the same environment in the department of Cusco, Peru, by molecular analysis, 265 faecal specimens were obtained from alpacas, llamas, sheep and shepherd children, and tested for RVA by RT-PCR. Genotyping was performed by multiplex PCR and sequence analysis. Rotavirus A was detected in 20.3% of alpaca, 47.5% of llama, 100% of sheep and 33.3% of human samples. The most common genetic constellations were G3-P[40]-I8-E3-H6 in alpacas, G1/G3-P[8]-I1-E1-H1 in llamas, G1/G3/G35-P[1]/P[8]-I1-E1-H1 in sheep and G3-P[40]-I1/I8-E3-H1 in humans. The newly described genotypes P[40] and P[50] were identified in all host species, including humans. Genotyping showed that the majority of samples presented coinfection with two or more RVA strains. These data demonstrate the great genetic diversity of RVA in animals and humans in Cusco, Peru. Phylogenetic analysis suggested that the strains represent zoonotic transmission among the species studied. Due to the characteristics of the human and animal populations in this study (cohabitation of different host species in conditions of poor sanitation and hygiene), the occurrence of zoonoses is a real possibility.
Collapse
Affiliation(s)
- Miguel Rojas
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratorio de Microbiologia y Parasitologia, Facultad de Medicina Veterinaria, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Helver G Dias
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge Luiz S Gonçalves
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alberto Manchego
- Laboratorio de Microbiologia y Parasitologia, Facultad de Medicina Veterinaria, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Raul Rosadio
- Laboratorio de Microbiologia y Parasitologia, Facultad de Medicina Veterinaria, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Danilo Pezo
- Instituto Veterinario de Investigaciones Tropicales y de Altura, Cusco, Peru
| | - Norma Santos
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
111
|
Chen S, Feng C, Fang Y, Zhou X, Xu L, Wang W, Kong X, P Peppelenbosch M, Pan Q, Yin Y. The Eukaryotic Translation Initiation Factor 4F Complex Restricts Rotavirus Infection via Regulating the Expression of IRF1 and IRF7. Int J Mol Sci 2019; 20:ijms20071580. [PMID: 30934842 PMCID: PMC6480131 DOI: 10.3390/ijms20071580] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
The eIF4F complex is a translation initiation factor that closely regulates translation in response to a multitude of environmental conditions including viral infection. How translation initiation factors regulate rotavirus infection remains poorly understood. In this study, the knockdown of the components of the eIF4F complex using shRNA and CRISPR/Cas9 were performed, respectively. We have demonstrated that loss-of-function of the three components of eIF4F, including eIF4A, eIF4E and eIF4G, remarkably promotes the levels of rotavirus genomic RNA and viral protein VP4. Consistently, knockdown of the negative regulator of eIF4F and programmed cell death protein 4 (PDCD4) inhibits the expression of viral mRNA and the VP4 protein. Mechanically, we confirmed that the silence of the eIF4F complex suppressed the protein level of IRF1 and IRF7 that exert potent antiviral effects against rotavirus infection. Thus, these results demonstrate that the eIF4F complex is an essential host factor restricting rotavirus replication, revealing new targets for the development of new antiviral strategies against rotavirus infection.
Collapse
Affiliation(s)
- Sunrui Chen
- Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China.
- Erasmus MC-University Medical Center, 3000 CA Rotterdam, The Netherlands.
| | - Cui Feng
- Department of Materials Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Yan Fang
- College of Basic Medicine, Shannxi University of Chinese Medicine, Xianyang 712046, China.
| | - Xinying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China.
| | - Lei Xu
- College of Life Sciences, Northwest A&F University, Yangling 712100, China.
| | - Wenshi Wang
- Erasmus MC-University Medical Center, 3000 CA Rotterdam, The Netherlands.
| | - Xiangdong Kong
- Department of Materials Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | | | - Qiuwei Pan
- Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China.
- Erasmus MC-University Medical Center, 3000 CA Rotterdam, The Netherlands.
| | - Yuebang Yin
- Erasmus MC-University Medical Center, 3000 CA Rotterdam, The Netherlands.
| |
Collapse
|
112
|
Abstract
BACKGROUND Rotavirus results in more diarrhoea-related deaths in children under five years 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. Rotavirus vaccines that have been prequalified by the World Health Organization (WHO) include a monovalent vaccine (RV1; Rotarix, GlaxoSmithKline), a pentavalent vaccine (RV5; RotaTeq, Merck), and, more recently, another monovalent vaccine (Rotavac, Bharat Biotech). OBJECTIVES To evaluate rotavirus vaccines prequalified by the WHO (RV1, RV5, and Rotavac) for their efficacy and safety in children. SEARCH METHODS On 4 April 2018 we searched MEDLINE (via PubMed), the Cochrane Infectious Diseases Group Specialized Register, CENTRAL (published in the Cochrane Library), Embase, LILACS, and BIOSIS. We also searched the WHO ICTRP, ClinicalTrials.gov, clinical trial reports from manufacturers' websites, and reference lists of included studies and relevant systematic reviews. SELECTION CRITERIA We selected randomized controlled trials (RCTs) in children comparing rotavirus vaccines prequalified for use by the WHO versus placebo or no intervention. DATA COLLECTION AND ANALYSIS Two review authors independently assessed trial eligibility and assessed risks of bias. One review author extracted data and a second author cross-checked them. We combined dichotomous data using the risk ratio (RR) and 95% confidence interval (CI). We stratified the analysis by country mortality rate and used GRADE to evaluate evidence certainty. MAIN RESULTS Fifty-five trials met the inclusion criteria and enrolled a total of 216,480 participants. Thirty-six trials (119,114 participants) assessed RV1, 15 trials (88,934 participants) RV5, and four trials (8432 participants) Rotavac.RV1 Children vaccinated and followed up the first year of life In low-mortality countries, RV1 prevents 84% of severe rotavirus diarrhoea cases (RR 0.16, 95% CI 0.09 to 0.26; 43,779 participants, 7 trials; high-certainty evidence), and probably prevents 41% of cases of severe all-cause diarrhoea (RR 0.59, 95% CI 0.47 to 0.74; 28,051 participants, 3 trials; moderate-certainty evidence). In high-mortality countries, RV1 prevents 63% of severe rotavirus diarrhoea cases (RR 0.37, 95% CI 0.23 to 0.60; 6114 participants, 3 trials; high-certainty evidence), and 27% of severe all-cause diarrhoea cases (RR 0.73, 95% CI 0.56 to 0.95; 5639 participants, 2 trials; high-certainty evidence).Children vaccinated and followed up for two yearsIn low-mortality countries, RV1 prevents 82% of severe rotavirus diarrhoea cases (RR 0.18, 95% CI 0.14 to 0.23; 36,002 participants, 9 trials; high-certainty evidence), and probably prevents 37% of severe all-cause diarrhoea episodes (rate ratio 0.63, 95% CI 0.56 to 0.71; 39,091 participants, 2 trials; moderate-certainty evidence). In high-mortality countries RV1 probably prevents 35% of severe rotavirus diarrhoea cases (RR 0.65, 95% CI 0.51 to 0.83; 13,768 participants, 2 trials; high-certainty evidence), and 17% of severe all-cause diarrhoea cases (RR 0.83, 95% CI 0.72 to 0.96; 2764 participants, 1 trial; moderate-certainty evidence).No increased risk of serious adverse events (SAE) was detected (RR 0.88 95% CI 0.83 to 0.93; high-certainty evidence). There were 30 cases of intussusception reported in 53,032 children after RV1 vaccination and 28 cases in 44,214 children after placebo or no intervention (RR 0.70, 95% CI 0.46 to 1.05; low-certainty evidence).RV5 Children vaccinated and followed up the first year of life In low-mortality countries, RV5 probably prevents 92% of severe rotavirus diarrhoea cases (RR 0.08, 95% CI 0.03 to 0.22; 4132 participants, 5 trials; moderate-certainty evidence). We did not identify studies reporting on severe all-cause diarrhoea in low-mortality countries. In high-mortality countries, RV5 prevents 57% of severe rotavirus diarrhoea (RR 0.43, 95% CI 0.29 to 0.62; 5916 participants, 2 trials; high-certainty evidence), but there is probably little or no difference between vaccine and placebo for severe all-cause diarrhoea (RR 0.80, 95% CI 0.58 to 1.11; 1 trial, 4085 participants; moderate-certainty evidence).Children vaccinated and followed up for two yearsIn low-mortality countries, RV5 prevents 82% of severe rotavirus diarrhoea cases (RR 0.18, 95% CI 0.08 to 0.39; 7318 participants, 4 trials; moderate-certainty evidence). We did not identify studies reporting on severe all-cause diarrhoea in low-mortality countries. In high-mortality countries, RV5 prevents 41% of severe rotavirus diarrhoea cases (RR 0.59, 95% CI 0.43 to 0.82; 5885 participants, 2 trials; high-certainty evidence), and 15% of severe all-cause diarrhoea cases (RR 0.85, 95% CI 0.75 to 0.98; 5977 participants, 2 trials; high-certainty evidence).No increased risk of serious adverse events (SAE) was detected (RR 0.93 95% CI 0.86 to 1.01; moderate to high-certainty evidence). There were 16 cases of intussusception in 43,629 children after RV5 vaccination and 20 cases in 41,866 children after placebo (RR 0.77, 95% CI 0.41 to 1.45; low-certainty evidence).Rotavac Children vaccinated and followed up the first year of life Rotavac has not been assessed in any RCT in countries with low child mortality. In India, a high-mortality country, Rotavac probably prevents 57% of severe rotavirus diarrhoea cases (RR 0.43, 95% CI 0.30 to 0.60; 6799 participants, moderate-certainty evidence); the trial did not report on severe all-cause diarrhoea at one-year follow-up.Children vaccinated and followed up for two yearsRotavac probably prevents 54% of severe rotavirus diarrhoea cases in India (RR 0.46, 95% CI 0.35 to 0.60; 6541 participants, 1 trial; moderate-certainty evidence), and 16% of severe all-cause diarrhoea cases (RR 0.84, 95% CI 0.71 to 0.98; 6799 participants, 1 trial; moderate-certainty evidence).No increased risk of serious adverse events (SAE) was detected (RR 0.93 95% CI 0.85 to 1.02; moderate-certainty evidence). There were eight cases of intussusception in 5764 children after Rotavac vaccination and three cases in 2818 children after placebo (RR 1.33, 95% CI 0.35 to 5.02; very low-certainty evidence).There was insufficient evidence of an effect on mortality from any rotavirus vaccine (198,381 participants, 44 trials; low- to very low-certainty evidence), as the trials were not powered to detect an effect at this endpoint. AUTHORS' CONCLUSIONS RV1, RV5, and Rotavac prevent episodes of rotavirus diarrhoea. Whilst the relative effect estimate is smaller in high-mortality than in low-mortality countries, there is a greater number of episodes prevented in these settings as the baseline risk is much higher. We found no increased risk of serious adverse events.
Collapse
Affiliation(s)
- Karla Soares‐Weiser
- CochraneEditorial & Methods DepartmentSt Albans House, 57 ‐ 59 HaymarketLondonUKSW1Y 4QX
| | - Hanna Bergman
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
| | - Nicholas Henschke
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
| | - Femi Pitan
- Chevron Corporation2 Chevron DriveLekkiLagosNigeria
| | - Nigel Cunliffe
- University of LiverpoolInstitute of Infection and Global Health, Faculty of Health and Life SciencesLiverpoolUKL69 7BE
| |
Collapse
|
113
|
Gunawan E, Utsumi T, Wahyuni RM, Dinana Z, Sudarmo SM, Shoji I, Soetjipto, Lusida MI. Post-vaccinated asymptomatic rotavirus infections: A community profile study of children in Surabaya, Indonesia. J Infect Public Health 2019; 12:625-629. [PMID: 30837151 DOI: 10.1016/j.jiph.2019.02.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/22/2019] [Accepted: 02/10/2019] [Indexed: 10/27/2022] Open
Abstract
BACKGROUND Rotavirus gastroenteritis accounts for significant childhood morbidity and mortality worldwide. Vaccination using RotarixTM (GSK) and RotaTeq® (Merck) was introduced due to the tremendous disease burden. The possibility of asymptomatic infections following vaccinations was poorly understood. This study examined rotavirus cases in post-vaccinated children, their clinical manifestations and the genotypes of isolated strains. METHODS Stool samples of healthy, vaccinated children under 5 years of age in Surabaya were collected monthly for 1 year between January 2016 and February 2017. Episodes of gastroenteritis were reported, and samples were collected. Rotavirus was identified using multiplex reverse transcription Polymerase Chain Reaction (QIAGEN, Inc., Valencia, CA). Clinical manifestations were measured using the Vesikari score. The genotype was analyzed by Applied Biosystems (Foster, CA). RESULTS A total of 109 stool samples were collected from 30 subjects, of which 22 received Rotarix; 8 RotaTeq. Nine out of 109 samples were collected during diarrhea episodes of 8 subjects. Two asymptomatic rotavirus infections were identified by RT-PCR. The genotypes isolated were G1P[8] and G3P[8]. CONCLUSIONS Asymptomatic rotavirus infections can occur in post-vaccinated children. Strains identified were homologous to serotypes eliciting gastroenteritis in unvaccinated children of the same community.
Collapse
Affiliation(s)
- Emily Gunawan
- Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Takako Utsumi
- Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia; Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Rury M Wahyuni
- Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Zayyin Dinana
- Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Subijanto M Sudarmo
- Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Department of Child Health, Dr. Soetomo General Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Ikuo Shoji
- Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Soetjipto
- Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia; Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia; Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Maria I Lusida
- Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia; Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia; Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
| |
Collapse
|
114
|
Gillard P, Tamura T, Kuroki H, Morikawa Y, Moerman L, Parra J, Kitamura Y, Mihara K, Okamasa A. Immunogenicity and safety of the diphtheria, pertussis, tetanus and inactivated poliovirus vaccine when co-administered with the human rotavirus vaccine (Rotarix) in healthy Japanese infants: a phase IV randomized study. Hum Vaccin Immunother 2019; 15:800-808. [PMID: 30785851 PMCID: PMC6605875 DOI: 10.1080/21645515.2018.1564441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Rotavirus infections have been reported to account for 40–50% of all hospitalized acute gastroenteritis cases in young children (<5 years) in Japan. Since 2011, Rotarix containing the live attenuated human rotavirus RIX4414 strain (HRV) has been licensed in Japan for infants. Vaccination against rotavirus is optional in Japan whereas administration of diphtheria, pertussis, tetanus, and inactivated poliovirus (DPT-IPV) vaccine is part of the national routine immunization program. In this open-label, randomized, controlled, multicenter study, we evaluated the immunogenicity and safety of the DPT-IPV vaccine (Squarekids) administered concomitantly or staggered with the liquid HRV (Rotarix) vaccine in healthy Japanese infants. A total of 292 infants aged 6–12 weeks were randomly assigned to receive DPT-IPV vaccine and HRV vaccine co-administered (n = 147) or staggered (n = 145). Immune responses to DPT-IPV vaccine were evaluated by measuring the post-vaccination serum antibody titers/concentrations to each antigen at one month following the third dose of DPT-IPV vaccine. Seroprotection/seropositivity against each of the diphtheria, pertussis (pertussis toxin and filamentous hemagglutinin), tetanus, and poliovirus type 1, 2 and 3 antigens was 92.8% or higher in both groups. In terms of immunogenicity, DPT-IPV vaccine co-administered with HRV vaccine was shown to be non-inferior to DPT-IPV vaccine with a staggered administration. The safety profile was comparable in the two vaccine groups with no vaccine-related serious adverse events, no deaths and no cases of intussusception. These results support co-administration of HRV vaccine with DPT-IPV vaccine in Japan. ClinicalTrials.gov NCT02907216
Collapse
Affiliation(s)
| | - Tsuyoshi Tamura
- b Department of Pediatrics , Hashimoto Clinic , Tokyo , Japan
| | - Haruo Kuroki
- c Department of Pediatrics , Sotobo Children's Clinic , Chiba , Japan
| | | | | | - Jose Parra
- f Clinical Statistics , GSK , Wavre , Belgium
| | - Yurina Kitamura
- g Project Management, Japan Development Division , GSK , Tokyo , Japan
| | - Kazuko Mihara
- h Medicines Development Division , GSK , Tokyo , Japan
| | - Arisa Okamasa
- h Medicines Development Division , GSK , Tokyo , Japan
| |
Collapse
|
115
|
Steele A, Victor J, Carey M, Tate J, Atherly D, Pecenka C, Diaz Z, Parashar U, Kirkwood C. Experiences with rotavirus vaccines: can we improve rotavirus vaccine impact in developing countries? Hum Vaccin Immunother 2019; 15:1215-1227. [PMID: 30735087 PMCID: PMC6663148 DOI: 10.1080/21645515.2018.1553593] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 11/06/2018] [Accepted: 11/22/2018] [Indexed: 12/16/2022] Open
Abstract
Rotavirus vaccines have been introduced into over 95 countries globally and demonstrate substantial impact in reducing diarrheal mortality and diarrheal hospitalizations in young children. The vaccines are also considered by WHO as "very cost effective" interventions for young children, particularly in countries with high diarrheal disease burden. Yet the full potential impact of rotavirus immunization is yet to be realized. Large countries with big birth cohorts and where disease burden is high in Africa and Asia have not yet implemented rotavirus vaccines at all or at scale. Significant advances have been made demonstrating the impact of the vaccines in low- and lower-middle income countries, yet the modest effectiveness of the vaccines in these settings is challenging. Current research highlights these challenges and considers alternative strategies to overcome them, including alternative immunization schedules and host factors that may inform us of new opportunities.
Collapse
Affiliation(s)
- A.D. Steele
- Enteric and Diarrheal Diseases, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - J.C. Victor
- Policy, Access and Innovation, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - M.E. Carey
- Enteric and Diarrheal Diseases, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - J.E. Tate
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - D.E. Atherly
- Policy, Access and Innovation, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - C. Pecenka
- Policy, Access and Innovation, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Z. Diaz
- Enteric and Diarrheal Diseases, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - U.D. Parashar
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - C.D. Kirkwood
- Enteric and Diarrheal Diseases, Bill & Melinda Gates Foundation, Seattle, WA, USA
| |
Collapse
|
116
|
Carvalho-Costa FA, de Assis RMS, Fialho AM, Araújo IT, Silva MF, Gómez MM, Andrade JS, Rose TL, Fumian TM, Volotão EM, Miagostovich MP, Leite JPG. The evolving epidemiology of rotavirus A infection in Brazil a decade after the introduction of universal vaccination with Rotarix®. BMC Pediatr 2019; 19:42. [PMID: 30704518 PMCID: PMC6354375 DOI: 10.1186/s12887-019-1415-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 01/22/2019] [Indexed: 01/15/2023] Open
Abstract
Background Brazil introduced the monovalent rotavirus vaccine (Rotarix®) in 2006. This study aimed to assess the epidemiology and genotype distribution of species-A rotavirus (RVA) in Brazil, comparing the pre- and post-vaccination periods. Methods Laboratory-based RVA surveillance included 866 municipalities in 22 Brazilian states, over a 21-year period. A total of 16,185 children with diarrheal diseases (DD) aged up to 12 years between 1996 and 2005 (pre-vaccination period, n = 7030) and from 2006 to 2017 (post-vaccination period, n = 9155) were enrolled. RVA was detected using ELISA immune assay and/or polyacrylamide gel electrophoresis and genotyped using nested PCR and/or nucleotide sequencing. RVA-positivity and genotypes detection rates were compared in distinct periods and age groups and Rotarix vaccination status. Results RVA-positivity in pre- and post-vaccination periods was, respectively: 4–11 months bracket, 33.3% (668/2006) and 16.3% (415/2547) (p < 0.001); 12–24 months, 28.2% (607/2154) and 22.2% (680/3068) (p < 0.001); 25–48 months, 17.4% (215/1235) and 29.4% (505/1720) (p < 0.001). Genotypes distribution in the pre- and post-vaccination periods was, respectively: G1P [8]/G1P[Not Typed], 417/855 (48.8%) and 118/1835 (6.4%) (p < 0.001); G2P [4]/G2P[NT], 47/855 (5.5%) and 838/1835 (45.7%) (p < 0.001); G3P [8]/G3P[NT], 55/855 (6.4%) and 253/1835 (13.8%) (p < 0.001); G9P [8]/G9P[NT], 238/855 (27.8%) and 152/1835 (8.3%) (p < 0.001); G12P [8]/G129P[NT], 0/871 (0%) and 249/1835(13.6%) (p < 0.001). Concerning infants aged 4–11 months, RVA frequency in fully vaccinated and non-vaccinated individuals was 11.9% (125/1052) and 24.5% (58/237) (p < 0.001), respectively. In children aged 12–24 months, RVA detection rate was 18.1% (253/1395) and 29.6% (77/260) (p < 0.001), for the vaccinated and non-vaccinated individuals, respectively (p < 0.001). Conclusions RVA infection was significantly less frequent in children aged ≤2 years with DD after implementing vaccination, mainly among vaccinated children. It was also observed a decrease of P [8] circulation and emergence of G2P[4] in 2005, and afterwards in the post-vaccine era, with spreading of G12P[8] in 2014–2015 and of G3P[8] in 2017. Continuous RVA surveillance must be carried out in this scenario.
Collapse
Affiliation(s)
- Filipe A Carvalho-Costa
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365 Pavilhão Hélio e Peggy Pereira, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil. .,Laboratory of Epidemiology and Molecular Systematics, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365 Pavilhão Leonidas Deane, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil. .,Regional Office Fiocruz Piauí. Rua Magalhães Filho, n° 519, Centro/Norte, Teresina, Piauí, Brazil.
| | - Rosane M S de Assis
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365 Pavilhão Hélio e Peggy Pereira, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alexandre M Fialho
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365 Pavilhão Hélio e Peggy Pereira, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Irene T Araújo
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365 Pavilhão Hélio e Peggy Pereira, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelle F Silva
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365 Pavilhão Hélio e Peggy Pereira, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariela M Gómez
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365 Pavilhão Hélio e Peggy Pereira, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana S Andrade
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365 Pavilhão Hélio e Peggy Pereira, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tatiana L Rose
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365 Pavilhão Hélio e Peggy Pereira, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tulio M Fumian
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365 Pavilhão Hélio e Peggy Pereira, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduardo M Volotão
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365 Pavilhão Hélio e Peggy Pereira, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marize P Miagostovich
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365 Pavilhão Hélio e Peggy Pereira, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil
| | - José Paulo G Leite
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365 Pavilhão Hélio e Peggy Pereira, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
117
|
Walker JL, Andrews NJ, Atchison CJ, Collins S, Allen DJ, Ramsay ME, Ladhani SN, Thomas SL. Effectiveness of oral rotavirus vaccination in England against rotavirus-confirmed and all-cause acute gastroenteritis. Vaccine X 2019; 1:100005. [PMID: 31384727 PMCID: PMC6668223 DOI: 10.1016/j.jvacx.2019.100005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 11/29/2022] Open
Abstract
Rotarix® had very good vaccine effectiveness (VE) in UK public health use. Two-dose VE against confirmed infection in young children was 77% (95%CI:66–85%) The vaccine programme was exceptionally successful (>90% vaccine uptake, high VE) Thus, it is highly likely that most acute gastroenteritis (AGE) was no longer due to rotavirus. This explains the lack of demonstrable VE against all-cause AGE.
Background The monovalent oral rotavirus vaccine Rotarix® was introduced into the UK infant immunisation programme in 2013. We estimated vaccine effectiveness (VE) in the first two years of the programme. Methods We used a test-negative case-control design and enhanced national surveillance data for 1869 vaccine-eligible children tested for rotavirus infection to obtain adjusted odds ratios and VE against laboratory-confirmed rotavirus infections. Linked anonymised UK primary care and hospitalisation data from the Clinical Practice Research Datalink (40,723 children) and random-effects Poisson regression were used in a cohort study to estimate VE against all-cause acute gastroenteritis (AGE) and AGE hospitalisations. Results VE against laboratory-confirmed infection was 69% (95% Confidence Interval: 40–84%) for one dose and 77% (95%CI: 66–85%) for two doses. Two-dose VE in children aged <12 months and ≥12 months was 85% (95%CI: 74–91%) and 54% (95%CI: 15–75%), respectively. In contrast, we found no evidence that the vaccine was effective against all-cause AGE (VE = −20%, 95%CI: −36% to −5%), or against AGE hospitalisations (VE = 35%, 95% CI: −86% to 77%). Conclusions In this first detailed assessment of VE of the Rotarix® vaccine in the English national programme, we show that Rotarix® was highly effective in preventing laboratory-confirmed rotavirus infection in young children. This provides reassurance about the vaccine’s performance in real-life settings and gives key information for future cost-effectiveness analyses. The high VE against rotavirus-specific AGE, and the exceptionally successful implementation of the national rotavirus vaccine programme (with >90% vaccine coverage), explains the lack of VE against all-cause AGE because most AGE in the post-vaccine era would not have been due to rotavirus, although some underestimation of VE could also have occurred due to differential healthcare utilisation by vaccinated and unvaccinated infants. This highlights the importance of using specific vaccine-preventable endpoints for these scenarios.
Collapse
Affiliation(s)
- Jemma L Walker
- Statistics, Modelling and Economics Department, Public Health England, 61 Colindale Avenue, London NW9 5EQ, UK.,Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Nick J Andrews
- Statistics, Modelling and Economics Department, Public Health England, 61 Colindale Avenue, London NW9 5EQ, UK.,Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Christina J Atchison
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Sarah Collins
- Immunisation and Countermeasures, Public Health England, 61 Colindale Avenue, London NW9 5EQ, UK
| | - David J Allen
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Mary E Ramsay
- Immunisation and Countermeasures, Public Health England, 61 Colindale Avenue, London NW9 5EQ, UK
| | - Shamez N Ladhani
- Immunisation and Countermeasures, Public Health England, 61 Colindale Avenue, London NW9 5EQ, UK
| | - Sara L Thomas
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
118
|
Yu J, Lai S, Geng Q, Ye C, Zhang Z, Zheng Y, Wang L, Duan Z, Zhang J, Wu S, Parashar U, Yang W, Liao Q, Li Z. Prevalence of rotavirus and rapid changes in circulating rotavirus strains among children with acute diarrhea in China, 2009-2015. J Infect 2019; 78:66-74. [PMID: 30017609 PMCID: PMC11373190 DOI: 10.1016/j.jinf.2018.07.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/13/2018] [Accepted: 07/01/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Rotavirus is a leading cause of morbidity and mortality in young children worldwide. In China, the universal immunization of children with the rotavirus vaccine has not been introduced, and the two globally distributed vaccines (RotaTeq and Rotarix) are not licensed in the country. We aim to determine the prevalence and strain diversity of rotavirus in children with diarrhea aged ≤ five years across China. MATERIALS AND METHODS Sentinel-based surveillance of acute diarrhea was conducted at 213 participating hospitals in China from January 1, 2009, through December 31, 2015. Group A rotavirus (RVA) was tested by using enzyme-linked immunosorbent assays, and G- and P-genotype of RVA were tested by RT-PCR methods. RESULTS Of 33,616 children with diarrhea, 10,089 (30%) were positive for RVA; RVA-associated diarrhea was identified in 2247 (39.5%, n = 2247/5685) inpatients and 7842 (28.1%, n = 7842/27931) outpatients. Children living in low-middle-income regions suffered from the highest burden of rotavirus, with 40.7% of diarrhea cases attributed to rotavirus infection, followed by 31.3% in upper-middle-income and 11.2% in high-income regions. The majority of children (88.9%, n = 8976/10089) who tested positive for RVA were children aged ≤ 2 years. The seasonal peak of RVA was in the winter. Among all 2533 RVA strains genotyped, five strain combinations, G9P[8], G3P[8], G1P[8], G2P[4] and G3P[4], contributed to 71.3% (1807/2533) of the RVA-associated diarrhea cases. The predominant strain of RVA has rapidly evolved from G3P[8] and G1P[8] to G9P[8] in the recent years, with the proportion of G9P[8] having increased remarkably from 3.4% in 2009 to 60.9% in 2015. CONCLUSIONS The burden of diarrhea attributed to rotavirus is high in China, highlighting the potential value of vaccination. The rapid shift of RVA strains highlights the importance of conducting rotavirus surveillance to ensure that currently marketed vaccines provide protective efficacy against the circulating strains.
Collapse
Affiliation(s)
- Jianxing Yu
- MOH Key Laboratory of Systems Biology of Pathogens and Dr. Christophe Mérieux Laboratory, CAMS-Fondation Mérieux, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, 9 Dongdan 3rd Alley, Dongcheng District, Beijing 100730, China; Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China
| | - Shengjie Lai
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China; WorldPop Department of Geography and Environment, University of Southampton, Southampton SO17 1BJ, UK; School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, 130 Dongan Road, Shanghai 200032, China.; Flowminder Foundation, Roslagsgatan 17, SE-11355 Stockholm, Sweden
| | - Qibin Geng
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China; State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Chuchu Ye
- Research Base of Key Laboratory of Surveillance and Early-warning on Infectious Disease in China CDC, Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China; School of Public Health, Fudan University, Shanghai 200032, China
| | - Zike Zhang
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yaming Zheng
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China
| | - Liping Wang
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China
| | - Zhaojun Duan
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China
| | - Jing Zhang
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China
| | - Shuyu Wu
- Division of Global Health Protection, Center for Global Health, United States Centers for Disease Control and Prevention, Beijing 100600, China
| | - Umesh Parashar
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia 30329, USA
| | - Weizhong Yang
- Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China
| | - Qiaohong Liao
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China.
| | - Zhongjie Li
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China.
| |
Collapse
|
119
|
Ogden KM, Tan Y, Akopov A, Stewart LS, McHenry R, Fonnesbeck CJ, Piya B, Carter MH, Fedorova NB, Halpin RA, Shilts MH, Edwards KM, Payne DC, Esona MD, Mijatovic-Rustempasic S, Chappell JD, Patton JT, Halasa NB, Das SR. Multiple Introductions and Antigenic Mismatch with Vaccines May Contribute to Increased Predominance of G12P[8] Rotaviruses in the United States. J Virol 2019; 93:e01476-18. [PMID: 30333170 PMCID: PMC6288334 DOI: 10.1128/jvi.01476-18] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/09/2018] [Indexed: 01/19/2023] Open
Abstract
Rotavirus is the leading global cause of diarrheal mortality for unvaccinated children under 5 years of age. The outer capsid of rotavirus virions consists of VP7 and VP4 proteins, which determine viral G and P types, respectively, and are primary targets of neutralizing antibodies. Successful vaccination depends upon generating broadly protective immune responses following exposure to rotaviruses presenting a limited number of G- and P-type antigens. Vaccine introduction resulted in decreased rotavirus disease burden but also coincided with the emergence of uncommon G and P genotypes, including G12. To gain insight into the recent predominance of G12P[8] rotaviruses in the United States, we evaluated 142 complete rotavirus genome sequences and metadata from 151 clinical specimens collected in Nashville, TN, from 2011 to 2013 through the New Vaccine Surveillance Network. Circulating G12P[8] strains were found to share many segments with other locally circulating strains but to have distinct constellations. Phylogenetic analyses of G12 sequences and their geographic sources provided evidence for multiple separate introductions of G12 segments into Nashville, TN. Antigenic epitopes of VP7 proteins of G12P[8] strains circulating in Nashville, TN, differ markedly from those of vaccine strains. Fully vaccinated children were found to be infected with G12P[8] strains more frequently than with other rotavirus genotypes. Multiple introductions and significant antigenic mismatch may in part explain the recent predominance of G12P[8] strains in the United States and emphasize the need for continued monitoring of rotavirus vaccine efficacy against emerging rotavirus genotypes.IMPORTANCE Rotavirus is an important cause of childhood diarrheal disease worldwide. Two immunodominant proteins of rotavirus, VP7 and VP4, determine G and P genotypes, respectively. Recently, G12P[8] rotaviruses have become increasingly predominant. By analyzing rotavirus genome sequences from stool specimens obtained in Nashville, TN, from 2011 to 2013 and globally circulating rotaviruses, we found evidence of multiple introductions of G12 genes into the area. Based on sequence polymorphisms, VP7 proteins of these viruses are predicted to present themselves to the immune system very differently than those of vaccine strains. Many of the sick children with G12P[8] rotavirus in their diarrheal stools also were fully vaccinated. Our findings emphasize the need for continued monitoring of circulating rotaviruses and the effectiveness of the vaccines against strains with emerging G and P genotypes.
Collapse
Affiliation(s)
- Kristen M Ogden
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yi Tan
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- J. Craig Venter Institute, Rockville, Maryland, USA
| | - Asmik Akopov
- J. Craig Venter Institute, Rockville, Maryland, USA
| | - Laura S Stewart
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Rendie McHenry
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Bhinnata Piya
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Maximilian H Carter
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | - Meghan H Shilts
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kathryn M Edwards
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Daniel C Payne
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Mathew D Esona
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - James D Chappell
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John T Patton
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Natasha B Halasa
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Suman R Das
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- J. Craig Venter Institute, Rockville, Maryland, USA
| |
Collapse
|
120
|
Vojtek I, Buchy P, Doherty TM, Hoet B. Would immunization be the same without cross-reactivity? Vaccine 2018; 37:539-549. [PMID: 30591255 DOI: 10.1016/j.vaccine.2018.12.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/07/2018] [Accepted: 12/04/2018] [Indexed: 01/08/2023]
Abstract
"Cross-reactivity" (the observed immune response against pathogen types not specifically targeted by the vaccine antigen composition) and "cross-protection" (clinical protection against related non-vaccine microorganism types) are vaccinology concepts that are attracting renewed interest in the context of disease prevention. National health authorities are collecting mounting evidence of the importance of cross-reactivity. For some vaccines, this has been substantiated by cross-protection data from clinical studies and/or post-licensure data, where their introduction into immunization programmes has shown beneficial impacts on disease caused by related non-vaccine microorganisms. This knowledge has influenced the way new vaccines are designed, developed, and evaluated in real-life settings. Some of the new vaccines are now designed with the specific aim of having a greater breadth of protection. Ideal vaccine antigens therefore include epitopes with conserved homology across related pathogen types, because it is not always possible to include the antigens of all the individual types of a given pathogen species. The use of novel adjuvants with greater immunostimulatory properties can also contribute to improved overall vaccine cross-reactivity, as could the use of antigen delivery platforms. The growing body of evidence allows us to better understand the full impact of vaccines - beyond vaccine-type disease - which should be taken into consideration when assessing the full value of vaccination programmes.
Collapse
Affiliation(s)
- Ivo Vojtek
- GSK, Avenue Fleming 20, 1300 Wavre, Belgium.
| | | | | | | |
Collapse
|
121
|
Willame C, Vonk Noordegraaf-Schouten M, Gvozdenović E, Kochems K, Oordt-Speets A, Praet N, van Hoorn R, Rosillon D. Effectiveness of the Oral Human Attenuated Rotavirus Vaccine: A Systematic Review and Meta-analysis-2006-2016. Open Forum Infect Dis 2018; 5:ofy292. [PMID: 30539038 PMCID: PMC6284461 DOI: 10.1093/ofid/ofy292] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 11/07/2018] [Indexed: 01/04/2023] Open
Abstract
Background Gastroenteritis caused by rotavirus accounts for considerable morbidity in young children. We aimed to assess the vaccine effectiveness (VE) of the oral rotavirus vaccine Rotarix, as measured by laboratory-confirmed rotavirus infection after referral to hospital and/or emergency departments in children aged <5 years with gastroenteritis. Methods We performed a systematic search for peer-reviewed studies conducted in real-life settings published between 2006 and 2016 and a meta-analysis to calculate the overall Rotarix VE, which was further discriminated through stratified analyses. Results The overall VE estimate was 69% (95% confidence interval [CI], 62% to 75%); stratified analyses revealed a non-negligible impact of factors such as study design and socioeconomic status. Depending on the control group, VE ranged from 63% (95% CI, 52% to 72%) to 81% (95% CI, 69% to 88%) for unmatched and matched rotavirus test–negative controls. VE varied with socioeconomic status: 81% (95% CI, 74% to 86%) in high-income countries, 54% (95% CI, 39% to 65%) in upper-middle-income countries, and 63% (95% CI, 50% to 72%) in lower-middle-income countries. Age, rotavirus strain, and disease severity were also shown to impact VE, but to a lesser extent. Conclusions This meta-analysis of real-world studies showed that Rotarix is effective in helping to prevent hospitalizations and/or emergency department visits due to rotavirus infection.
Collapse
Affiliation(s)
| | | | | | - Katrin Kochems
- Pallas Health Research and Consultancy, Rotterdam, the Netherlands
| | | | | | - Rosa van Hoorn
- Pallas Health Research and Consultancy, Rotterdam, the Netherlands
| | | |
Collapse
|
122
|
Muendo C, Laving A, Kumar R, Osano B, Egondi T, Njuguna P. Prevalence of rotavirus infection among children with acute diarrhoea after rotavirus vaccine introduction in Kenya, a hospital cross-sectional study. BMC Pediatr 2018; 18:323. [PMID: 30309343 PMCID: PMC6180366 DOI: 10.1186/s12887-018-1291-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 09/26/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Rotavirus infection is the most common cause of acute gastroenteritis globally in children under 5 years of age and is responsible for approximately 5% of all child deaths yearly. Rotavirus vaccination is considered an effective public health strategy to prevent infection and reduce the severity of disease. Multi-centre country trials on rotavirus vaccines demonstrated efficacy rates of more than 85% in developed countries but only about 65% in developing nations. Rotavirus vaccination was introduced into the Kenya Expanded Programme on Immunization (KEPI) in 2014. The objective of our study was to determine the prevalence of rotavirus infection, severity of acute diarrhoea and to determine the rotavirus vaccination status among children aged 3-24 months presenting with acute diarrhoea at Kenyatta National Hospital after introduction of rotavirus vaccine in Kenya. METHODS A total of 365 children aged 3-24 months presenting with acute diarrhoea at KNH were recruited from August 2016 to April 2017. Data on rotavirus vaccination status, nutritional status, feeding practices and sociodemographic characteristics were obtained and a full clinical evaluation of the patients was done. Severity of the gastroenteritis was assessed using the 20 point Vesikari Clinical Severity Scoring System. The children who were admitted were followed up for 7 days using hospital ward registers. Comorbid conditions were established from patient's clinical records and physical examination. Stool specimens from study participants were tested for rotavirus using a commercially available enzyme linked immunosorbent immunoassay kit- ProSpecT Rotavirus Microplate Assay. RESULTS Majority of the children (96.7%) had received rotavirus vaccinations. The overall rotavirus prevalence was 14.5% and was higher among 17-24 months at 19.5%. The prevalence somewhat differed by gender, nutritional status, exclusive breastfeeding status, age and education level of mother/caregiver. Overall, a half of the children had severe acute diarrhoea and there were some differences in severity by child/mother characteristics. CONCLUSION There is still burden of rotavirus diarrhoea after introduction of rotavirus vaccine and the prevalence varies by child characteristics.
Collapse
Affiliation(s)
| | - Ahmed Laving
- Department of Paediatrics and Child Health, University of Nairobi, P.O. Box 19676–00202, Nairobi, Kenya
| | - Rashmi Kumar
- Department of Paediatrics and Child Health, University of Nairobi, P.O. Box 19676–00202, Nairobi, Kenya
| | - Boniface Osano
- Department of Paediatrics and Child Health, University of Nairobi, P.O. Box 19676–00202, Nairobi, Kenya
| | - Thaddaeus Egondi
- Drugs for Neglected Diseases initiative, P.O. Box 21936–00505, Nairobi, Kenya
| | - Pamela Njuguna
- Public Health Specialist, Afya Resource Associates, P. O. Box 238–00202, Nairobi, Kenya
| |
Collapse
|
123
|
Abebe A, Getahun M, Mapaseka SL, Beyene B, Assefa E, Teshome B, Tefera M, Kebede F, Habtamu A, Haile-Mariam T, Jeffrey Mphahlele M, Teshager F, Ademe A, Teka T, Weldegebriel GG, Mwenda JM. Impact of rotavirus vaccine introduction and genotypic characteristics of rotavirus strains in children less than 5 years of age with gastroenteritis in Ethiopia: 2011-2016. Vaccine 2018; 36:7043-7047. [PMID: 30301641 DOI: 10.1016/j.vaccine.2018.09.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/05/2018] [Accepted: 09/21/2018] [Indexed: 10/28/2022]
Abstract
INTRODUCTION A monovalent rotavirus vaccine was introduced in the Ethiopian Expanded Program on Immunization from November 2013. We compared impact of rotavirus vaccine introduction on rotavirus associated acute diarrhea hospitalizations and genotypic characteristics of rotavirus strains pre-and post-vaccine introduction. METHODS Sentinel surveillance for diarrhea among children <5 years of age was conducted at 3 hospitals in Addis Ababa, Ethiopia from 2011 to 2017. Stool specimens were collected from enrolled children and tested using an antigen capture enzyme immunoassay. Rotavirus positive samples (156 from pre- and 141 from post-vaccination periods) were further characterized by rotavirus genotyping methods to identify the predominant G and P types circulating during the surveillance era. RESULTS A total of 788 children were enrolled during the pre- (July 2011-June 2013) and 815 children during the post-vaccination (July 2014-June 2017) periods. The proportion of diarrhea hospitalizations due to rotavirus among children <5 years of age declined by 17% from 24% (188/788) in the pre-vaccine period and to 20% (161/185) in post-vaccine introduction era. Similarly, a reduction of 18% in proportion of diarrhea hospitalizations due to rotavirus in children <12 months of age in the post (27%) vs pre-vaccine (33%) periods was observed. Seasonal peaks of rotavirus declined following rotavirus vaccine introduction. The most prevalent circulating strains were G12P[8] in 2011 (36%) and in 2012 (27%), G2P[4] (35%) in 2013, G9P[8] (19%) in 2014, G3P[6] and G2P[4] (19% each) in 2015, and G3P[8] (29%) in 2016. DISCUSSION Following rotavirus vaccine introduction in Ethiopia, a reduction in rotavirus associated hospitalizations was seen in all age groups with the greatest burden in children <12 months of age. A wide variety of rotavirus strains circulated in the pre- and post-vaccine introduction periods.
Collapse
Affiliation(s)
- Almaz Abebe
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia.
| | | | - Seheri L Mapaseka
- SAMRC Diarrhoeal Pathogens Research Unit, Department of Virology, Sefako Makgatho Health Sciences University, Medunsa, Pretoria, South Africa
| | - Berhane Beyene
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Essete Assefa
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Birke Teshome
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Mesfin Tefera
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | | | - Abebe Habtamu
- Black Lion Hospital, AAU Medical Faculty, Addis Ababa, Ethiopia
| | | | - M Jeffrey Mphahlele
- SAMRC Diarrhoeal Pathogens Research Unit, Department of Virology, Sefako Makgatho Health Sciences University, Medunsa, Pretoria, South Africa
| | | | | | - Telahun Teka
- Yekatit 12 Hospital, AAU Medical Faculty, Addis Ababa, Ethiopia
| | | | - Jason M Mwenda
- WHO Regional Office for Africa (WHO/AFRO), Brazzaville, People's Republic of Congo
| |
Collapse
|
124
|
Ledent E, Arlegui H, Buyse H, Basile P, Karkada N, Praet N, Nachbaur G. Benefit Versus Risk Assessment of Rotavirus Vaccination in France: A Simulation and Modeling Analysis. BioDrugs 2018; 32:139-152. [PMID: 29589230 PMCID: PMC5878204 DOI: 10.1007/s40259-018-0273-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction Two vaccines against rotavirus gastroenteritis (RVGE) in young children, Rotarix and RotaTeq, have been available in Europe since 2006. Vaccination against rotaviruses significantly reduces the burden of RVGE, but it is also associated with a very small increased risk of intussusception. In a benefit–risk analysis, the prevented RVGE burden is weighed against the possible excess of intussusception. Purpose The aim was to compare the estimated benefits and risks of Rotarix vaccination in France. Methods We estimated the benefits (vaccine-preventable RVGE hospitalizations and deaths) and risks (vaccine-caused intussusception hospitalizations and deaths) following two doses of Rotarix in a birth cohort of 791,183 followed for 3–5 years in France. We used data from peer-reviewed clinical and epidemiological studies or publications, and government statistics. Results Within the total number of French children below 5 years of age, we estimate vaccination could prevent a median 11,132 [95% credible interval (CI) 7842–14,408] RVGE hospitalizations and 7.43 (95% CI 3.27–14.68) RVGE deaths. At the same time, vaccination could cause an average of 6.86 (95% CI 2.25–38.37) intussusception hospitalizations and 0.0099 (95% CI 0.0024–0.060) intussusception deaths in the entire French birth cohort of infants below 1 year of age. Therefore, for every intussusception hospitalization and every intussusception death caused by vaccination, 1624 (95% CI 240–5243) RVGE hospitalizations and 743 (95% CI 93–3723) RVGE deaths are prevented, respectively, by vaccination. Conclusions The vaccine-prevented RVGE hospitalizations and deaths (benefit) greatly outweigh the excess potentially vaccination-related cases of intussusception (risk), indicating a favorable benefit–risk balance for Rotarix in France. Electronic supplementary material The online version of this article (10.1007/s40259-018-0273-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Hugo Arlegui
- Clinical Research, NPI and Academic Alliances, Laboratoire GSK, Rueil-Malmaison, France
| | - Hubert Buyse
- Clinical Safety and Pharmacovigilance, GSK, Wavre, Belgium
| | - Peter Basile
- Clinical Safety and Pharmacovigilance, GSK, Wavre, Belgium
| | | | - Nicolas Praet
- Clinical Research and Development, GSK, Wavre, Belgium
| | - Gaëlle Nachbaur
- Pharmaco-Epidemiology and Health Outcomes Research, Laboratoire GSK, Rueil-Malmaison, France
| |
Collapse
|
125
|
Carvalho MF, Gill D. Rotavirus vaccine efficacy: current status and areas for improvement. Hum Vaccin Immunother 2018; 15:1237-1250. [PMID: 30215578 PMCID: PMC6663136 DOI: 10.1080/21645515.2018.1520583] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/12/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022] Open
Abstract
The difference noted in Rotavirus vaccine efficiency between high and low income countries correlates with the lack of universal access to clean water and higher standards of hygiene. Overcoming these obstacles will require great investment and also time, therefore more effective vaccines should be developed to meet the needs of those who would benefit the most from them. Increasing our current knowledge of mucosal immunity, response to Rotavirus infection and its modulation by circadian rhythms could point at actionable pathways to improve vaccination efficacy, especially in the case of individuals affected by environmental enteropathy. Also, a better understanding and validation of Rotavirus entry factors as well as the systematic monitoring of dominant strains could assist in tailoring vaccines to individual's needs. Another aspect that could improve vaccine efficiency is targeting to M cells, for which new ligands could potentially be sought. Finally, alternative mucosal adjuvants and vaccine expression, storage and delivery systems could have a positive impact in the outcome of Rotavirus vaccination.
Collapse
Affiliation(s)
| | - Davinder Gill
- MSD Wellcome Trust Hilleman Laboratories Pvt. Ltd., New Delhi, India
| |
Collapse
|
126
|
Sadiq A, Bostan N, Yinda KC, Naseem S, Sattar S. Rotavirus: Genetics, pathogenesis and vaccine advances. Rev Med Virol 2018; 28:e2003. [PMID: 30156344 DOI: 10.1002/rmv.2003] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 07/02/2018] [Accepted: 07/07/2018] [Indexed: 01/27/2023]
Abstract
Since its discovery 40 years ago, rotavirus (RV) is considered to be a major cause of infant and childhood morbidity and mortality particularly in developing countries. Nearly every child in the world under 5 years of age is at the risk of RV infection. It is estimated that 90% of RV-associated mortalities occur in developing countries of Africa and Asia. Two live oral vaccines, RotaTeq (RV5, Merck) and Rotarix (RV1, GlaxoSmithKline) have been successfully deployed to scale down the disease burden in Europe and America, but they are less effective in Africa and Asia. In April 2009, the World Health Organization recommended the inclusion of RV vaccination in national immunization programs of all countries with great emphasis in developing countries. To date, 86 countries have included RV vaccines into their national immunization programs including 41 Global Alliance for Vaccines and Immunization eligible countries. The predominant RV genotypes circulating all over the world are G1P[8], G2P[4], G3P[8], G4P[8], and G9P[8], while G12[P6] and G12[P8] are emerging genotypes. On account of the segmented genome, RV shows an enormous genetic diversity that leads to the evolution of new genotypes that can influence the efficacy of current vaccines. The current need is for a global RV surveillance program to monitor the prevalence and antigenic variability of new genotypes to formulate future vaccine development planning. In this review, we will summarize the previous and recent insights into RV structure, classification, and epidemiology and current status of RV vaccination around the globe and will also cover the status of RV research and vaccine policy in Pakistan.
Collapse
Affiliation(s)
- Asma Sadiq
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Nazish Bostan
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Kwe Claude Yinda
- Rega Institute, Laboratory of Clinical and Epidemiological Virology, University of Leuven, Leuven, Belgium
| | - Saadia Naseem
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Sadia Sattar
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| |
Collapse
|
127
|
Yin Y, Chen S, Hakim MS, Wang W, Xu L, Dang W, Qu C, Verhaar AP, Su J, Fuhler GM, Peppelenbosch MP, Pan Q. 6-Thioguanine inhibits rotavirus replication through suppression of Rac1 GDP/GTP cycling. Antiviral Res 2018; 156:92-101. [PMID: 29920300 PMCID: PMC7113846 DOI: 10.1016/j.antiviral.2018.06.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 06/11/2018] [Accepted: 06/15/2018] [Indexed: 02/06/2023]
Abstract
Rotavirus infection has emerged as an important cause of complications in organ transplantation recipients and might play a role in the pathogenesis of inflammatory bowel disease (IBD). 6-Thioguanine (6-TG) has been widely used as an immunosuppressive drug for organ recipients and treatment of IBD in the clinic. This study aims to investigate the effects and mode-of-action of 6-TG on rotavirus replication. Human intestinal Caco2 cell line, 3D model of human primary intestinal organoids, laboratory rotavirus strain (SA11) and patient-derived rotavirus isolates were used. We have demonstrated that 6-TG significantly inhibits rotavirus replication in these intestinal epithelium models. Importantly, gene knockdown or knockout of Rac1, the cellular target of 6-TG, significantly inhibited rotavirus replication, indicating the supportive role of Rac1 for rotavirus infection. We have further demonstrated that 6-TG can effectively inhibit the active form of Rac1 (GTP-Rac1), which essentially mediates the anti-rotavirus effect of 6-TG. Consistently, ectopic over-expression of GTP-Rac1 facilitates but an inactive Rac1 (N17) or a specific Rac1 inhibitor (NSC23766) inhibits rotavirus replication. In conclusion, we have identified 6-TG as an effective inhibitor of rotavirus replication via the inhibition of Rac1 activation. Thus, for transplantation patients or IBD patients infected with rotavirus or at risk of rotavirus infection, the choice of 6-TG as a treatment appears rational. 6-TG inhibits rotavirus infection. Rac1, the cellular target of 6-TG, supports rotavirus infection. 6-TG inhibits the active form of Rac1 (GTP-Rac1) to exert the anti-rotavirus effect.
Collapse
Affiliation(s)
- Yuebang Yin
- Center for Biomedical Research, Northwest Minzu University, Lanzhou, China; Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands; Institute of Subtropical Agriculture, Chinese Academic of Sciences, Changsha 410125, China
| | - Sunrui Chen
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Mohamad S Hakim
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands; Department of Microbiology, Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Wenshi Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Lei Xu
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Wen Dang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Changbo Qu
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Auke P Verhaar
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Junhong Su
- Center for Biomedical Research, Northwest Minzu University, Lanzhou, China; Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands; Medical Faculty, Kunming University of Science and Technology, Kunming, PR China
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Qiuwei Pan
- Center for Biomedical Research, Northwest Minzu University, Lanzhou, China; Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
128
|
Abstract
Two vaccines available for protection against rotavirus gastroenteritis (RVGE), Rotarix and RotaTeq, have contributed to a large decrease in the incidence of paediatric diarrhoea in countries where they have been used. However, they have also led to a small increase in the risk of intussusception. Methods: We compare the number of prevented hospitalisations for RVGE to the number of vaccine-induced hospitalised intussusceptions in France. Results: With 9.5% coverage (French 2015 estimation), vaccination was estimated to prevent, annually, a median of 1,074 hospitalisations (2.5th and 97.5th percentiles (2.5th–97.5th): 810–1,378) and 1.4 deaths (2.5th–97.5th: 1.2–1.6) from RVGE. It was also estimated to cause, annually, 5.0 hospitalisations (2.5th–97.5th: 3.2–7.7) and 0.005 deaths (2.5th–97.5th: 0.001–0.015) from intussusception. The benefit–risk ratio is therefore 214 (2.5th–97.5th: 128–362) for hospitalisations and 273 (2.5th–97.5th: 89–1,228) for deaths. Under a hypothetical 92% coverage, rotavirus vaccination with Rotarix would avoid 10,459 (2.5th–97.5th: 7,702–13,498) hospitalisations for RVGE and induce 47.0 (2.5th–97.5th: 25.1–81.4) hospitalisations for intussusception annually, thereby preventing 13.7 (2.5th–97.5th: 11.1–15.2) deaths and inducing 0.05 (2.5th–97.5th: 0.01–0.15) deaths. Conclusion: The benefit–risk ratio in France is similar to that of other European countries.
Collapse
Affiliation(s)
- Adnane Lamrani
- Biostatistics, Biomathematics, Pharmacoepidemiology and Infectious Diseases (B2PHI), Inserm, UVSQ, Institut Pasteur, Université Paris-Saclay, Villejuif, France
| | - Pascale Tubert-Bitter
- Biostatistics, Biomathematics, Pharmacoepidemiology and Infectious Diseases (B2PHI), Inserm, UVSQ, Institut Pasteur, Université Paris-Saclay, Villejuif, France
| | - Catherine Hill
- Centre de Recherche en Epidémiologie et Santé des Population (CESP), Inserm, Villejuif, France
| | - Sylvie Escolano
- Biostatistics, Biomathematics, Pharmacoepidemiology and Infectious Diseases (B2PHI), Inserm, UVSQ, Institut Pasteur, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
129
|
Jonesteller CL, Burnett E, Yen C, Tate JE, Parashar UD. Effectiveness of Rotavirus Vaccination: A Systematic Review of the First Decade of Global Postlicensure Data, 2006-2016. Clin Infect Dis 2018; 65:840-850. [PMID: 28444323 DOI: 10.1093/cid/cix369] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/18/2017] [Indexed: 11/13/2022] Open
Abstract
Two rotavirus vaccines, Rotarix (RV1) and RotaTeq (RV5), were licensed for global use in 2006. A systematic review of 48 peer- reviewed articles with postlicensure data from 24 countries showed a median RV1 vaccine effectiveness (VE) of 84%, 75%, and 57% in countries with low, medium, and high child mortality, respectively, and RV5 VE of 90% and 45% in countries with low and high child mortality, respectively. A partial vaccine series provided considerable protection, but not to the same level as a full series. VE tended to decline in the second year of life, particularly in medium- and high-mortality settings, and tended to be greater against more severe rotavirus disease. Postlicensure data from countries across geographic regions and with different child mortality levels demonstrate that under routine use, both RV1 and RV5 are effective against rotavirus disease, supporting the World Health Organization recommendation that all countries introduce rotavirus vaccine into their national immunization program.
Collapse
Affiliation(s)
| | - Eleanor Burnett
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Catherine Yen
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | | |
Collapse
|
130
|
Mwanza-Lisulo M, Chomba MS, Chama M, Besa EC, Funjika E, Zyambo K, Banda R, Imikendu M, Sianongo S, Hancock REW, Lee A, Chilengi R, Stagg AJ, Namangala B, Kelly PM. Retinoic acid elicits a coordinated expression of gut homing markers on T lymphocytes of Zambian men receiving oral Vivotif, but not Rotarix, Dukoral or OPVERO vaccines. Vaccine 2018; 36:4134-4141. [PMID: 29801999 PMCID: PMC6020133 DOI: 10.1016/j.vaccine.2018.04.083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/29/2018] [Accepted: 04/30/2018] [Indexed: 11/28/2022]
Abstract
ATRA increased vaccine specific IgA in gut secretions to Vivotif but not Dukoral or Rotarix. ATRA increased α4β7 and CCR9 gut marker expression in a coordinated manner only when given simultaneously with Vivotif vaccine. In individuals with coordinated gut marker expression Vivotif specific IgA increase was much stronger.
All-trans retinoic acid (ATRA) up-regulates, in laboratory animals, the expression of the gut homing markers α4β7 integrin and CCR9 on lymphocytes, increasing their gut tropism. Here, we show that, in healthy adult volunteers, ATRA induced an increase of these gut homing markers on T cells in vivo in a time dependent manner. The coordinated increase of α4β7 and CCR9 by ATRA was seen in 57% (12/21) of volunteers and only when given together with an oral Vivotif vaccine. When this coordinated response to ATRA and Vivotif vaccine was present, it was strongly correlated with the gut immunoglobulin A (IgA) specific response to vaccine LPS (ρ = 0.82; P = 0.02). Using RNA-Seq analysis of whole blood transcription, patients receiving ATRA and Vivotif in conjunction showed transcriptomic changes in immune-related pathways, particularly including interferon α/β signaling pathway, membrane-ECM interactions and immune hubs. These results suggest that exogenous ATRA can be used to manipulate responses to a subclass of oral vaccines, so far limited to a live attenuated Vivotif vaccine.
Collapse
Affiliation(s)
- Mpala Mwanza-Lisulo
- Tropical Gastroenterology & Nutrition Group, Department of Medicine, School of Medicine, University of Zambia, Lusaka, Zambia.
| | - Mumba S Chomba
- Tropical Gastroenterology & Nutrition Group, Department of Medicine, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Mubanga Chama
- Tropical Gastroenterology & Nutrition Group, Department of Medicine, School of Medicine, University of Zambia, Lusaka, Zambia; Department of Chemistry, University of Zambia, Lusaka, Zambia
| | - Ellen C Besa
- Tropical Gastroenterology & Nutrition Group, Department of Medicine, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Evelyn Funjika
- Tropical Gastroenterology & Nutrition Group, Department of Medicine, School of Medicine, University of Zambia, Lusaka, Zambia; Department of Chemistry, University of Zambia, Lusaka, Zambia
| | - Kanekwa Zyambo
- Tropical Gastroenterology & Nutrition Group, Department of Medicine, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Rose Banda
- Tropical Gastroenterology & Nutrition Group, Department of Medicine, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Mercy Imikendu
- Tropical Gastroenterology & Nutrition Group, Department of Medicine, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Sandie Sianongo
- Tropical Gastroenterology & Nutrition Group, Department of Medicine, School of Medicine, University of Zambia, Lusaka, Zambia
| | | | - Amy Lee
- University of British Columbia, Vancouver, Canada
| | - Roma Chilengi
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka, Zambia
| | - Andy J Stagg
- Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, UK
| | | | - Paul M Kelly
- Tropical Gastroenterology & Nutrition Group, Department of Medicine, School of Medicine, University of Zambia, Lusaka, Zambia; Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, UK
| |
Collapse
|
131
|
Magwira CA, Taylor MB. Composition of gut microbiota and its influence on the immunogenicity of oral rotavirus vaccines. Vaccine 2018; 36:3427-3433. [PMID: 29752022 DOI: 10.1016/j.vaccine.2018.04.091] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/09/2018] [Accepted: 04/24/2018] [Indexed: 02/07/2023]
Abstract
The introduction of oral rotavirus vaccines (ORVVs) has led to a reduction in number of hospitalisations and deaths due to rotavirus (RV) infection. However, the efficacy of the vaccines has been varied with low-income countries showing significantly lower efficacy as compared to high-income countries. The reasons for the disparity are not fully understood but are thought to be multi-factorial. In this review article, we discuss the concept that the disparity in the efficacy of oral rotavirus vaccines between the higher and lower socio-economical countries could be due the nature of the bacteria that colonises and establishes in the gut early in life. We further discuss recent studies that has demonstrated significant correlations between the composition of the gut bacteria and the immunogenicity of oral vaccines, and their implications in the development of novel oral RV vaccines or redesigning the current ones for maximum impact.
Collapse
Affiliation(s)
- Cliff A Magwira
- Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, South Africa.
| | - Maureen B Taylor
- Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, South Africa; School of Health Systems and Public Health, Faculty of Health Sciences, University of Pretoria, South Africa
| |
Collapse
|
132
|
High prevalence of G3 rotavirus in hospitalized children in Rawalpindi, Pakistan during 2014. PLoS One 2018; 13:e0195947. [PMID: 29708975 PMCID: PMC5927433 DOI: 10.1371/journal.pone.0195947] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/03/2018] [Indexed: 12/23/2022] Open
Abstract
Rotavirus A species (RVA) is the leading cause of severe diarrhea among children in both developed and developing countries. Among different RVA G types, humans are most commonly infected with G1, G2, G3, G4 and G9. During 2003-2004, G3 rotavirus termed as "new variant G3" emerged in Japan that later disseminated to multiple countries across the world. Although G3 rotaviruses are now commonly detected globally, they have been rarely reported from Pakistan. We investigated the genetic diversity of G3 strains responsible RVA gastroenteritis in children hospitalized in Rawalpindi, Pakistan during 2014. G3P[8] (18.3%; n = 24) was detected as the most common genotype causing majority of infections in children less than 06 months. Phylogenetic analysis of Pakistani G3 strains showed high amino acid similarity to "new variant G3" and G3 strains reported from China, Russia, USA, Japan, Belgium and Hungary during 2007-2012. Pakistani G3 strains belonged to lineage 3 within sub-lineage 3d, containing an extra N-linked glycosylation site compared to the G3 strain of RotaTeqTM. To our knowledge, this is the first report on the molecular epidemiology of G3 rotavirus strains from Pakistan and calls for immediate response measures to introduce RV vaccine in the routine immunization program of the country on priority.
Collapse
|
133
|
Arnold MM. Rotavirus vaccines: why continued investment in research is necessary. CURRENT CLINICAL MICROBIOLOGY REPORTS 2018; 5:73-81. [PMID: 29805958 PMCID: PMC5967271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
PURPOSE OF REVIEW Rotavirus vaccines were first introduced more than a decade ago and have had a tremendous impact on reducing the number of hospitalizations and deaths due to rotavirus-associated diarrhea. This review will discuss current rotavirus vaccines, post-licensure surveillance, progress in non-replicating vaccine development, and why continued research is important for understanding a virus that remains a globally leading cause of death due to diarrhea. RECENT FINDINGS Research advances have enhanced our understanding of how vaccines induce protection against subsequent severe disease, how the virus replicates and spreads in the face of the host immune system, and basic mechanisms governing the viral life cycle. SUMMARY Much remains to be learned about how to improve vaccine success, what are the molecular determinants of host range and virulence, and what are the interactions of the virus with the host that drive its replicative success, among many other important questions.
Collapse
Affiliation(s)
- Michelle M. Arnold
- Corresponding author: Michelle M. Arnold, , Telephone: 318-675-4731, ORCID: 0000-0001-9219-3097
| |
Collapse
|
134
|
Abstract
Throughout human history, pathogens transmitted through feces from person to person have caused substantial mortality. Over a century ago civil engineers in high income countries developed approaches to collect and remove feces from the environment that in communities with high incomes and strong governments markedly reduced the burden of enteric disease. These approaches, however, have not been successfully extended to impoverished communities in low income countries. Water/sanitation/hygiene professionals have attempted, with only limited success, to reduce fecal exposure and human disease in the absence of definitive civil engineering approaches. Medical professionals have worked to develop vaccines against some of the most important fecal oral pathogens. Each of these approaches needs further development and adaptation to optimally address the burden of fecally transmitted diseases in impoverished communities.
Collapse
|
135
|
Arnold MM. Rotavirus Vaccines: Why Continued Investment in Research Is Necessary. CURRENT CLINICAL MICROBIOLOGY REPORTS 2018. [DOI: 10.1007/s40588-018-0079-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
136
|
Sarker AR, Sultana M, Mahumud RA, Van Der Meer R, Morton A. Cost-effectiveness analysis of introducing universal childhood rotavirus vaccination in Bangladesh. Hum Vaccin Immunother 2018; 14:189-198. [PMID: 29099653 PMCID: PMC5791570 DOI: 10.1080/21645515.2017.1356962] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/21/2017] [Accepted: 07/14/2017] [Indexed: 10/18/2022] Open
Abstract
Diarrhea is one of the world's leading killers of children, and globally, rotavirus is the most common cause of severe diarrhea among under 5 children. In Bangladesh, rotavirus kills nearly 6,000 under 5 children in each year. To reduce the burden of childhood rotavirus diseases, universal rotavirus vaccination is recommended by World Health Organization. The objective of this study is to assess the cost-effectiveness of introducing universal childhood rotavirus vaccination with the newly developed ROTAVAC vaccine in national Expanded Programme of Immunization in Bangladesh. We developed a decision model to examine the potential impact of vaccination in Bangladesh and to examine the effect if the vaccination is applied in the nationwide immunization program schedule. Introduction of childhood universal rotavirus vaccination in Bangladesh scenario appears as highly cost-effective and would offer substantial future benefits for the young population if vaccinated today. The cost per DALY averted of introducing the rotavirus vaccine compared with status quo is approximately US$ 740.27 and US$ 728.67 per DALY averted from the health system and societal perspective respectively which is "very cost-effective" using GDP threshold level according to World Health Organization definition. The results of this analysis seek to contribute to an evidence-based recommendation about the introduction of universal rotavirus vaccination in national Expanded Programme of Immunization (EPI) in Bangladesh.
Collapse
Affiliation(s)
- Abdur Razzaque Sarker
- Health Economics and Financing Research, Health Systems & Population Studies Division, ICDDR,B, Dhaka, Bangladesh
- Department of Management Science, University of Strathclyde, Glasgow, UK
| | - Marufa Sultana
- Health Economics and Financing Research, Health Systems & Population Studies Division, ICDDR,B, Dhaka, Bangladesh
| | - Rashidul Alam Mahumud
- Health Economics and Financing Research, Health Systems & Population Studies Division, ICDDR,B, Dhaka, Bangladesh
| | | | - Alec Morton
- Department of Management Science, University of Strathclyde, Glasgow, UK
| |
Collapse
|
137
|
|
138
|
Parker EPK, Ramani S, Lopman BA, Church JA, Iturriza-Gómara M, Prendergast AJ, Grassly NC. Causes of impaired oral vaccine efficacy in developing countries. Future Microbiol 2018; 13:97-118. [PMID: 29218997 PMCID: PMC7026772 DOI: 10.2217/fmb-2017-0128] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/13/2017] [Indexed: 12/12/2022] Open
Abstract
Oral vaccines are less immunogenic when given to infants in low-income compared with high-income countries, limiting their potential public health impact. Here, we review factors that might contribute to this phenomenon, including transplacental antibodies, breastfeeding, histo blood group antigens, enteric pathogens, malnutrition, microbiota dysbiosis and environmental enteropathy. We highlight several clear risk factors for vaccine failure, such as the inhibitory effect of enteroviruses on oral poliovirus vaccine. We also highlight the ambiguous and at times contradictory nature of the available evidence, which undoubtedly reflects the complex and interconnected nature of the factors involved. Mechanisms responsible for diminished immunogenicity may be specific to each oral vaccine. Interventions aiming to improve vaccine performance may need to reflect the diversity of these mechanisms.
Collapse
Affiliation(s)
- Edward PK Parker
- Department of Infectious Disease Epidemiology, St Mary's Campus, Imperial College London, London, W2 1PG, UK
| | | | - Benjamin A Lopman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - James A Church
- Centre for Paediatrics, Blizard Institute, Queen Mary University of London, London, E1 2AT, UK
| | - Miren Iturriza-Gómara
- Centre for Global Vaccine Research, Institute of Infection & Global Health, University of Liverpool, Liverpool, L69 7BE, UK
| | - Andrew J Prendergast
- Centre for Paediatrics, Blizard Institute, Queen Mary University of London, London, E1 2AT, UK
| | - Nicholas C Grassly
- Department of Infectious Disease Epidemiology, St Mary's Campus, Imperial College London, London, W2 1PG, UK
| |
Collapse
|
139
|
Hemming-Harlo M, Vesikari T, Uhari M, Renko M, Salminen M, Torcel-Pagnon L, Hartwig S, Simondon F, Bricout H. Sustained High Effectiveness of RotaTeq on Hospitalizations Attributable to Rotavirus-Associated Gastroenteritis During 4 Years in Finland. J Pediatric Infect Dis Soc 2017; 6:317-323. [PMID: 27760800 PMCID: PMC7107484 DOI: 10.1093/jpids/piw061] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/25/2016] [Indexed: 12/28/2022]
Abstract
KEY POINTS The effectiveness of pentavalent rotavirus vaccine against rotavirus-associated hospitalization was more than 90% 4 years after introduction into the national immunization program in Finland. A major impact on hospitalization for all-cause gastroenteritis was observed also. BACKGROUND Rotavirus vaccination with exclusive use of RotaTeq was added to the National Immunization Programme (NIP) of Finland in September 2009. The objective of our study was to estimate the effectiveness and impact of RotaTeq after 4 years of follow-up. METHODS Between 2009 and 2013, we conducted a prospective surveillance study of children aged <16 years with acute gastroenteritis (AGE) and admitted in 2 hospitals in Finland. Rotavirus and other gastroenteritis viruses were detected in stool samples by reverse-transcription polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assays. The effectiveness of RotaTeq was investigated by using a case-control design; wild-type rotavirus-positive children were classified as "cases" and rotavirus-negative children as "controls." Hospital discharge records were used to estimate the impact of RotaTeq on rotavirus-associated AGE (RV-AGE) and all-cause AGE (AC-AGE) hospitalizations of age-eligible children in the NIP by comparing the prevaccination (2001-2006) and post-NIP seasons (2009-2013). RESULTS The crude estimate of the effectiveness of RotaTeq to prevent RV-AGE hospitalization in NIP age-eligible children was 94.4% (95% confidence interval, 79.8%-98.4%). No change in prevalent wild-type rotavirus genotypes was observed. Vaccine-derived rotaviruses were detected in 8% of the children with RV-AGE, with a probable causal association in 2 children. Hospital discharge records revealed that RV-AGE and AC-AGE hospitalizations in children aged <16 years decreased in the two post-NIP seasons by 79% and 58%, respectively, compared to those in the prevaccination seasons. CONCLUSIONS Over 4 years of follow-up, high rotavirus vaccine coverage in the NIP (>95%) has led to a major reduction in RV-AGE and AC-AGE hospitalizations without a resurgence of rotavirus activity. However, rotavirus continues to circulate in older unvaccinated children.
Collapse
Affiliation(s)
- Maria Hemming-Harlo
- Vaccine Research Center, School of Medicine, University of Tampere,Corresponding Author: Maria Hemming-Harlo, MD, PhD, Biokatu 10, 33520 Tampere, Finland. E-mail:
| | - Timo Vesikari
- Vaccine Research Center, School of Medicine, University of Tampere
| | - Matti Uhari
- PEDEGO Research Unit, University of Oulu, Finland
| | - Marjo Renko
- PEDEGO Research Unit, University of Oulu, Finland
| | - Marjo Salminen
- Vaccine Research Center, School of Medicine, University of Tampere
| | | | | | - Francois Simondon
- IRD UMR 216, Faculté de Pharmacie, Université Paris Descartes, France
| | | |
Collapse
|
140
|
Serologic response to porcine circovirus type 1 (PCV1) in infants vaccinated with the human rotavirus vaccine, Rotarix™: A retrospective laboratory analysis. Hum Vaccin Immunother 2017; 13:237-244. [PMID: 27657348 PMCID: PMC5287324 DOI: 10.1080/21645515.2016.1231262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In 2010, porcine circovirus type 1 (PCV1) material was unexpectedly detected in the oral live-attenuated human rotavirus (RV) vaccine, Rotarix™ (GSK Vaccines, Belgium). An initial study (NCT01511133) found no immunologic response against PCV1 in 40 vaccinated infants. As a follow-up, the current study (NCT02153333), searched for evidence of post-vaccination serologic response to PCV1 in a larger number of archived serum samples. Unlike the previous study, serum anti-PCV1 antibodies were assessed with an adapted Immuno Peroxidase Monolayer Assay (IPMA) using a Vero-adapted PCV1 strain. Samples from 596 infants who participated in clinical trials of the human RV vaccine were randomly selected and analyzed. The observed anti-PCV1 antibody seropositivity rate 1–2 months post-dose 2 was approximately 1% [90% Confidence Interval (CI): 0.3–2.6] (3/299 samples) in infants who received the human RV vaccine and 0.3% [90% CI: 0.0–1.6] (1/297 samples) in those who received placebo; the difference between the groups was −0.66 [90% CI: −2.16–0.60]. One subject in the vaccinated group was also seropositive before vaccination. Notably, the seropositivity rate observed in vaccinated subjects was below that observed during assay qualification in samples from unvaccinated subjects outside of this study (2.5%; 5/200 samples). No serious adverse events had been reported in any of the 4 subjects providing anti-PCV1 positive samples during the 31-day post-vaccination follow-up period in the original studies. In conclusion, the presence of PCV1 in the human RV vaccine is considered to be a manufacturing quality issue and does not appear to pose a safety risk to vaccinated infants.
Collapse
|
141
|
Leino T, Baum U, Scott P, Ollgren J, Salo H. Impact of five years of rotavirus vaccination in Finland – And the associated cost savings in secondary healthcare. Vaccine 2017; 35:5611-5617. [DOI: 10.1016/j.vaccine.2017.08.052] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/18/2017] [Accepted: 08/20/2017] [Indexed: 11/30/2022]
|
142
|
Harris V, Ali A, Fuentes S, Korpela K, Kazi M, Tate J, Parashar U, Wiersinga WJ, Giaquinto C, de Weerth C, de Vos WM. Rotavirus vaccine response correlates with the infant gut microbiota composition in Pakistan. Gut Microbes 2017; 9:93-101. [PMID: 28891751 PMCID: PMC5989807 DOI: 10.1080/19490976.2017.1376162] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Rotavirus (RV) is the leading cause of diarrhea-related death in children worldwide and ninety-five percent of rotavirus deaths occur in Africa and Asia. Rotavirus vaccines (RVV) can dramatically reduce RV deaths, but have low efficacy in low-income settings where they are most needed. The intestinal microbiome may contribute to this decreased RVV efficacy. This pilot study hypothesizes that infants' intestinal microbiota composition correlates with RVV immune responses and that RVV responders have different gut microbiota as compared to non-responders. We conducted a nested, matched case-control study comparing the pre-vaccination intestinal microbiota composition between 10 6-week old Pakistani RVV-responders, 10 6-week old Pakistani RVV non-responders, and 10 healthy Dutch infants. RVV response was defined as an Immunoglobulin A of ≥20 IU/mL following Rotarix™(RV1) vaccination in an infant with a pre-vaccination IgA<20. Infants were matched in a 1:1 ratio using ranked variables: RV1 dosing schedule (6/10/14; 6/10; or 10/14 weeks), RV season, delivery mode, delivery place, breastfeeding practices, age and gender. Fecal microbiota analysis was performed using a highly reproducible phylogenetic microarray. RV1 response correlated with a higher relative abundance of bacteria belonging to Clostridium cluster XI and Proteobacteria, including bacteria related to Serratia and Escherichia coli. Remarkably, abundance of these Proteobacteria was also significantly higher in Dutch infants when compared to RV1-non-responders in Pakistan. This small but carefully matched study showed the intestinal microbiota composition to correlate with RV1 seroconversion in Pakistan infants, identifying signatures shared with healthy Dutch infants.
Collapse
Affiliation(s)
- Vanessa Harris
- Amsterdam Institute for Global Health and Development and Department of Global Health, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands,Center for Experimental and Molecular Medicine, Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands,CONTACT Vanessa Harris , Amsterdam Institute for Global Health and Development and Department of Global Health, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Asad Ali
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Susana Fuentes
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Katri Korpela
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands,RPU Immunobiology, Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Momin Kazi
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Jacqueline Tate
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Center for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Umesh Parashar
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Center for Disease Control and Prevention, Atlanta, Georgia, USA
| | - W. Joost Wiersinga
- Center for Experimental and Molecular Medicine, Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlo Giaquinto
- Department of Paediatrics, University of Padova, Padova, Italy
| | - Carolina de Weerth
- Behavioral Science Institute, Department of Developmental Psychology, Radboud University, Nijmegen, The Netherlands
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands,RPU Immunobiology, Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
143
|
Rahajamanana VL, Raboba JL, Rakotozanany A, Razafindraibe NJ, Andriatahirintsoa EJPR, Razafindrakoto AC, Mioramalala SA, Razaiarimanga C, Weldegebriel GG, Burnett E, Mwenda JM, Seheri M, Mphahlele MJ, Robinson AL. Impact of rotavirus vaccine on all-cause diarrhea and rotavirus hospitalizations in Madagascar. Vaccine 2017; 36:7198-7204. [PMID: 28958809 DOI: 10.1016/j.vaccine.2017.08.091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/09/2017] [Accepted: 08/16/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND Rotavirus vaccine was introduced into the Extended Program on Immunization in Madagascar in May 2014. We analyzed trends in prevalence of all cause diarrhea and rotavirus hospitalization in children <5years of age before and after vaccine introduction and assessed trend of circulating rotavirus genotypes at Centre Hospitalier Universitaire Mère Enfant Tsaralalàna (CHU MET). METHODS From January 2010 to December 2016, we reviewed the admission logbook to observe the rate of hospitalization caused by gastroenteritis among 19619 children <5years of age admitted at the hospital. In June 2013-December 2016, active rotavirus surveillance was also conducted at CHUMET with support from WHO. Rotavirus antigen was detected by EIA from stool specimen of children who are eligible for rotavirus gastroenteritis surveillance at sentinel site laboratory and rotavirus positive specimens were further genotyped at Regional Reference Laboratory by RT-PCR. RESULTS Diarrhea hospitalizations decreased after rotavirus vaccine introduction. The median proportion of annual hospitalizations due to diarrhea was 26% (range: 31-22%) before vaccine introduction; the proportion was 25% the year of vaccine introduction, 17% in 2015 and 16% in 2016. Rotavirus positivity paralleled patterns observed in diarrhea. Before vaccine introduction, 56% of stool specimens tested positive for rotavirus; the percent positive was 13% in 2015, 12% in 2016. Diverse genotypes were detected in the pre-vaccine period; the most common were G3P[8] (n=53; 66%), G2P[4] (n=12; 15%), and G1P[8] (n=11; 14%). 6 distinct genotypes were found in 2015; the most common genotype was G2P[4] (n=10; 67%), the remaining, 5, G12[P8], G3[P8], G1G3[P4], G3G12[P4][P8] and G1G3[NT] had one positive specimen each. CONCLUSIONS Following rotavirus vaccine introduction all-cause diarrhea and rotavirus-specific hospitalizations declined dramatically. The most common genotypes detected in the pre-vaccine period were G3P[8] and G2P[4] in 2015, the post vaccine period.
Collapse
Affiliation(s)
- V L Rahajamanana
- Department of Child Health, Teaching Hospital, Centre Hospitalier Universitaire Mère Enfant Tsaralàlana, Antananarivo, Madagascar.
| | - J L Raboba
- Department of Child Health, Teaching Hospital, Centre Hospitalier Universitaire Mère Enfant Tsaralàlana, Antananarivo, Madagascar
| | - A Rakotozanany
- Department of Child Health, Teaching Hospital, Centre Hospitalier Universitaire Mère Enfant Tsaralàlana, Antananarivo, Madagascar
| | - N J Razafindraibe
- Teaching Hospital, Centre Hospitalier Universitaire Andohatapenaka, Antananarivo, Madagascar
| | | | - A C Razafindrakoto
- Department of Child Health, Teaching Hospital, Centre Hospitalier Universitaire Mère Enfant Tsaralàlana, Antananarivo, Madagascar
| | - S A Mioramalala
- National Malaria Country Program, Public Health Ministry, Antananarivo, Madagascar
| | | | - G G Weldegebriel
- WHO Inter-Country Support Team: East and Southern Africa (WHO IST/ESA), Harare, Zimbabwe
| | - E Burnett
- Foundation for the Centers for Disease Control and Prevention, Division of Viral Diseases, Atlanta, USA
| | - J M Mwenda
- World Health Organization (WHO) Regional Office for Africa (WHO/AFRO), Brazzaville, Congo
| | - M Seheri
- Regional Rotavirus Reference Laboratory, SAMRC/Diarrheal Pathogens Research Unit, Department of Virology, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| | - M J Mphahlele
- Regional Rotavirus Reference Laboratory, SAMRC/Diarrheal Pathogens Research Unit, Department of Virology, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| | - A L Robinson
- Department of Child Health, Teaching Hospital, Centre Hospitalier Universitaire Mère Enfant Tsaralàlana, Antananarivo, Madagascar
| |
Collapse
|
144
|
Standaert B, Schecroun N, Ethgen O, Topachevskyi O, Morioka Y, Van Vlaenderen I. Optimising the introduction of multiple childhood vaccines in Japan: A model proposing the introduction sequence achieving the highest health gains. Health Policy 2017; 121:1303-1312. [PMID: 29079394 DOI: 10.1016/j.healthpol.2017.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 08/01/2017] [Accepted: 08/24/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND Many countries struggle with the prioritisation of introducing new vaccines because of budget limitations and lack of focus on public health goals. A model has been developed that defines how specific health goals can be optimised through immunisation within vaccination budget constraints. METHODS Japan, as a country example, could introduce 4 new pediatric vaccines targeting influenza, rotavirus, pneumococcal disease and mumps with known burden of disease, vaccine efficacies and maximum achievable coverages. Operating under budget constraints, the Portfolio-model for the Management of Vaccines (PMV) identifies the optimal vaccine ranking and combination for achieving the maximum QALY gain over a period of 10 calendar years in children <5 years old. This vaccine strategy, of interest and helpful for a healthcare decision maker, is compared with an unranked vaccine selection process. RESULTS Results indicate that the maximum QALY gain with a fixed annual vaccination budget of 500 billion Japanese Yen over a 10-year period is 72,288 QALYs using the optimal sequence of vaccine introduction (mumps [1st], followed by influenza [2nd], rotavirus [3rd], and pneumococcal [4th]). With exactly the same budget but without vaccine ranking, the total QALY gain can be 20% lower. CONCLUSION The PMV model could be a helpful tool for decision makers in those environments with limited budget where vaccines have to be selected for trying to optimise specific health goals.
Collapse
|
145
|
Al-Aidaroos AY, Standaert B, Meszaros K, Shibl AM. Economic assessment of rotavirus vaccination in Saudi Arabia. J Infect Public Health 2017; 10:564-571. [DOI: 10.1016/j.jiph.2016.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 10/16/2016] [Accepted: 11/18/2016] [Indexed: 01/03/2023] Open
|
146
|
Ndombo PK, Ndze VN, Fokunang C, Ashukem TN, Boula A, Kinkela MN, Ndode CE, Seheri ML, Bowen MD, Waku-Kouomou D, Esona MD. Pre-vaccine circulating group a rotavirus strains in under 5 years children with acute diarrhea during 1999-2013 in Cameroon. Virology 2017; 1. [PMID: 29051924 PMCID: PMC5645035 DOI: 10.15761/vrr.1000120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The aim of this review was to assess all the studies on rotavirus G and P characterization during the pre-vaccine period (1999-2013) in Cameroon to have a better basis for post-vaccine introduction evaluations. A retrospective study was done through a comprehensive review of published (PubMed, Google Scholar) and accessible unpublished data on rotavirus G and P genotypes circulating in five regions of Cameroon. Descriptive data were expressed as frequencies tables and proportions. A total of 1844 rotavirus positive cases were analyzed. In all, 1534 strains were characterized for the P (VP4) specificity. Six different VP4 genotypes were observed, including P [4], P [6], P [8], P [9], P [10] and P [14]. The most predominant P genotypes were P [8] at 42.6%, and P [6] at 37.9%. Mixed infections were observed at 5.3%, whereas 4.1% of the strains were P non-typeable. A total of 1518 rotavirus strains were characterized for the G (VP7) specificity. VP7 genotypes G1, G2, G3, G4, G5, G6, G8, G9, G10 and G12 were observed. G1 (35.3%), G3 (19.5%), G2 (14.9%) and G12 (10.1%) were the predominant G genotypes while G5 and G10 were least prevalent at 0.06% each. Approximately 5.1% of all strains were G non-typeable whereas 5.3% were mixed G genotypes. A total of 1472 strains were characterized for both G and P genes, from which 38 different G-P combinations were observed. Overall, G1P [8] (22%) was identified as the predominant rotavirus strain circulating in Cameroon followed by G3P [6] (15%). In conclusion, we observed that the genotypes identified in Cameroon during 1999-2013 were partially covered by the two WHO recommended rotavirus vaccines. This review provides comprehensive up-to-date information on rotavirus strain surveillance in Cameroon during the pre-vaccination era.
Collapse
Affiliation(s)
- Paul Koki Ndombo
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaounde, Cameroon, South Africa.,Rotavirus National Reference Laboratory, Mother and Child Centre of the Chantal Biya Foundation, Yaoundé, Cameroon, South Africa
| | - Valantine N Ndze
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaounde, Cameroon, South Africa.,Rotavirus National Reference Laboratory, Mother and Child Centre of the Chantal Biya Foundation, Yaoundé, Cameroon, South Africa
| | - Charles Fokunang
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaounde, Cameroon, South Africa
| | - Taku Nadesh Ashukem
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaounde, Cameroon, South Africa
| | - Angeline Boula
- Rotavirus National Reference Laboratory, Mother and Child Centre of the Chantal Biya Foundation, Yaoundé, Cameroon, South Africa
| | - Mina N Kinkela
- Rotavirus National Reference Laboratory, Mother and Child Centre of the Chantal Biya Foundation, Yaoundé, Cameroon, South Africa
| | - Corlins E Ndode
- Rotavirus National Reference Laboratory, Mother and Child Centre of the Chantal Biya Foundation, Yaoundé, Cameroon, South Africa
| | - Mapaseka L Seheri
- South Africa Medical Research Council/Diarrhoeal Pathogen Research Unit, Department of Virology, Faculty of health Sciences, Sefako Makgatho Health Sciences University, Medunsa, Pretoria, South Africa
| | - Michael D Bowen
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Mathew D Esona
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
147
|
Holmgren J, Parashar UD, Plotkin S, Louis J, Ng SP, Desauziers E, Picot V, Saadatian-Elahi M. Correlates of protection for enteric vaccines. Vaccine 2017; 35:3355-3363. [PMID: 28504192 PMCID: PMC11342448 DOI: 10.1016/j.vaccine.2017.05.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 04/04/2017] [Accepted: 05/03/2017] [Indexed: 12/26/2022]
Abstract
An immunological Correlate of Protection (CoP) is an immune response that is statistically interrelated with protection. Identification of CoPs for enteric vaccines would help design studies to improve vaccine performance of licensed vaccines in low income settings, and would facilitate the testing of future vaccines in development that might be more affordable. CoPs are lacking today for most existing and investigational enteric vaccines. In order to share the latest information on CoPs for enteric vaccines and to discuss novel approaches to correlate mucosal immune responses in humans with protection, the Foundation Mérieux organized an international conference of experts where potential CoPs for vaccines were examined using case-studies for both bacterial and viral enteric pathogens. Experts on the panel concluded that to date, all established enteric vaccine CoPs, such as those for hepatitis A, Vi typhoid and poliovirus vaccines, are based on serological immune responses even though these may poorly reflect the relevant gut immune responses or predict protective efficacy. Known CoPs for cholera, norovirus and rotavirus could be considered as acceptable for comparisons of similarly composed vaccines while more work is still needed to establish CoPs for the remaining enteric pathogens and their candidate vaccines. Novel approaches to correlate human mucosal immune responses with protection include the investigation of gut-originating antibody-secreting cells (ASCs), B memory cells and follicular helper T cells from samples of peripheral blood during their recirculation.
Collapse
Affiliation(s)
- Jan Holmgren
- University of Gothenburg Vaccine Research Institute, Box 435, S-40530 Gothenburg, Sweden.
| | - Umesh D Parashar
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta GA, United States.
| | - Stanley Plotkin
- University of Pennsylvania and Vaxconsult, LLC, United States.
| | - Jacques Louis
- Fondation Mérieux, 17 rue Bourgelat, 69002 Lyon, France.
| | - Su-Peing Ng
- Sanofi Pasteur, Global Medical Affairs, 2 Avenue du Pont Pasteur, 69367 Lyon cedex 07, France.
| | - Eric Desauziers
- Sanofi Pasteur, Global Medical Affairs, 2 Avenue du Pont Pasteur, 69367 Lyon cedex 07, France.
| | | | - Mitra Saadatian-Elahi
- Hospices Civils de Lyon, Groupement Hospitalier Edouard Herriot, 5 Place d'Arsonval, 69437 Lyon cedex 03, France.
| |
Collapse
|
148
|
Kotirum S, Vutipongsatorn N, Kongpakwattana K, Hutubessy R, Chaiyakunapruk N. Global economic evaluations of rotavirus vaccines: A systematic review. Vaccine 2017; 35:3364-3386. [PMID: 28504193 DOI: 10.1016/j.vaccine.2017.04.051] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 04/17/2017] [Accepted: 04/19/2017] [Indexed: 11/26/2022]
Abstract
INTRODUCTION World Health Organization (WHO) recommends Rotavirus vaccines to prevent and control rotavirus infections. Economic evaluations (EE) have been considered to support decision making of national policy. Summarizing global experience of the economic value of rotavirus vaccines is crucial in order to encourage global WHO recommendations for vaccine uptake. Therefore, a systematic review of economic evaluations of rotavirus vaccine was conducted. METHODS We searched Medline, Embase, NHS EED, EconLit, CEA Registry, SciELO, LILACS, CABI-Global Health Database, Popline, World Bank - e-Library, and WHOLIS. Full economic evaluations studies, published from inception to November 2015, evaluating Rotavirus vaccines preventing Rotavirus infections were included. The methods, assumptions, results and conclusions of the included studies were extracted and appraised using WHO guide for standardization of EE of immunization programs. RESULTS 104 relevant studies were included. The majority of studies were conducted in high-income countries. Cost-utility analysis was mostly reported in many studies using incremental cost-effectiveness ratio per DALY averted or QALY gained. Incremental cost per QALY gained was used in many studies from high-income countries. Mass routine vaccination against rotavirus provided the ICERs ranging from cost-saving to highly cost-effective in comparison to no vaccination among low-income countries. Among middle-income countries, vaccination offered the ICERs ranging from cost-saving to cost-effective. Due to low- or no subsidized price of rotavirus vaccines from external funders, being not cost-effective was reported in some high-income settings. CONCLUSION Mass vaccination against rotavirus was generally found to be cost-effective, particularly in low- and middle-income settings according to the external subsidization of vaccine price. On the other hand, it may not be a cost-effective intervention at market price in some high-income settings. This systematic review provides supporting information to health policy-makers and health professionals when considering rotavirus vaccination as a national program.
Collapse
Affiliation(s)
- Surachai Kotirum
- School of Pharmacy, Monash University Malaysia, Selangor, Malaysia; Social and Administrative Pharmacy Department, Faculty of Pharmacy, Rangsit University, Muang, Pathumthani, Thailand
| | - Naaon Vutipongsatorn
- School of Pharmacy, Monash University Malaysia, Selangor, Malaysia; Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | | | - Raymond Hutubessy
- World Health Organization, Initiative for Vaccine Research, Geneva, Switzerland
| | - Nathorn Chaiyakunapruk
- School of Pharmacy, Monash University Malaysia, Selangor, Malaysia; Center of Pharmaceutical Outcomes Research (CPOR), Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand; School of Pharmacy, University of Wisconsin, Madison, USA; Health and Well-being Cluster, Global Asia in the 21(st) Centuary (GA21) Platform, Monash University Malaysia, Selangor, Malaysia.
| |
Collapse
|
149
|
Yandle Z, Coughlan S, Drew RJ, O’Flaherty N, O’Gorman J, De Gascun C. Circulating rotavirus genotypes in the Irish paediatric population prior to the introduction of the vaccination programme. Ir J Med Sci 2017; 186:1003-1007. [DOI: 10.1007/s11845-017-1604-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/24/2017] [Indexed: 02/07/2023]
|
150
|
Mohanty E, Dwibedi B, Kar SK, Acharya AS. Epidemiological features and genetic characterization of virus strains in rotavirus associated gastroenteritis in children of Odisha in Eastern India. INFECTION GENETICS AND EVOLUTION 2017; 53:77-84. [PMID: 28438670 DOI: 10.1016/j.meegid.2017.04.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/22/2017] [Accepted: 04/14/2017] [Indexed: 12/18/2022]
Abstract
We have studied the clinical characteristics, severity and seasonality of rotavirus infection and prevalent genotypes in 652 non-rota vaccinated children in Odisha in eastern India. P genotypes were analysed for their association with host blood group antigens. P type of the virus is determined by the VP8* gene, and specific recognition of A - type of Histo - blood group antigen by P[14]VP8* has been reported. VP4, VP7 and VP6 genes of commonly identified G1P[8] strain were compared with genes of the same strain isolated from other parts of India, elsewhere and strains used for Rotarix and Rotateq vaccines. In 54.75% of children with gastroenteritis, rota virus was found. 9.65% of children had moderate, 78.07% severe, and 12.28% very severe disease as assessed using the Vesikari scoring system. The incidence of infection was highest during winter months. There was no association between any blood group and specific P genotypes. G1P[8] was the commonest cause of gastroenteritis, followed by G1P[11], G3P[8], G9P[8], G2P[4], G2P[6], G9P[4], G9P[11] and G1P[6]. Predominant G genotypes identified were G1 (72.9%), G9 (10.81%), G2 (8.10%) and G3 (8.10%). Sequence analysis of the VP7 gene, placed the G1P[8] strain in lineage 1 and of VP6 gene placed nine G1P[8] strains in subgroup II and one in subgroup I. The VP7 gene segment of two Odisha G1P[8] strains were found to cluster relatively close to the VP7 sequences of Rotarix vaccine. Antigenic differences were found with vaccine strains. Ten G1P[8] strains sequenced for the VP4 gene had 91-93% nucleotide and 92-96% amino acid identity with Rotateq vaccine P[8]). Rotarix vaccine VP4 had 89-91% nucleotide and 90-92% amino acid identity. Our findings indicate genetic variability of rotavirus strains circulating in the region and are significant, given the introduction of rota vaccination in the State.
Collapse
Affiliation(s)
- Eileena Mohanty
- Viral Diagnostic and Research Laboratory, Regional Medical Research Centre, Indian Council of Medical Research, Bhubaneswar 751023, Odisha, India
| | - Bhagirathi Dwibedi
- Viral Diagnostic and Research Laboratory, Regional Medical Research Centre, Indian Council of Medical Research, Bhubaneswar 751023, Odisha, India.
| | - S K Kar
- Viral Diagnostic and Research Laboratory, Regional Medical Research Centre, Indian Council of Medical Research, Bhubaneswar 751023, Odisha, India
| | - A S Acharya
- Viral Diagnostic and Research Laboratory, Regional Medical Research Centre, Indian Council of Medical Research, Bhubaneswar 751023, Odisha, India
| |
Collapse
|