1
|
Lin X, Li H. Diverse processes in rotavirus vaccine development. Hum Vaccin Immunother 2025; 21:2475609. [PMID: 40126359 PMCID: PMC11934161 DOI: 10.1080/21645515.2025.2475609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
Rotavirus is a major cause of severe diarrhea and mortality in children under five years of age, leading to approximately 128,500 deaths annually.1-3 Vaccination is the most effective strategy for preventing rotavirus infection. While two widely used vaccines, Rotarix and RotaTeq, have shown good efficacy in high-income countries, their effectiveness is lower in low- and middle-income countries due to factors such as malnutrition and poor sanitation.4-6 These challenges include complex vaccination schedules and high production costs. Researchers are working on novel vaccines, including inactivated virus and viral protein-based options, as well as virus-like particles and recombinant proteins.7-9 Improving vaccine stability and applicability is crucial for resource-limited settings, and global vaccination strategies are expected to significantly reduce infection burdens, improve child health, and contribute to the achievement of global health goals.10-14.
Collapse
Affiliation(s)
- Xiaochen Lin
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan Provincial Key Laboratory of Vaccine R&D for Major Infectious Diseases, Kunming, China
| | - Hongjun Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan Provincial Key Laboratory of Vaccine R&D for Major Infectious Diseases, Kunming, China
| |
Collapse
|
2
|
Wenger C, Asare EO, Kwon J, Li X, Mwinjiwa E, Chinkhumba J, Jere KC, Hungerford D, Cunliffe NA, Paltiel AD, Pitzer VE. Cost-effectiveness analysis of alternative infant and neonatal rotavirus vaccination schedules in Malawi. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0004341. [PMID: 40209158 PMCID: PMC11984971 DOI: 10.1371/journal.pgph.0004341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/28/2025] [Indexed: 04/12/2025]
Abstract
Rotavirus is the leading cause of severe diarrhea among children under five worldwide, especially in low- and middle-income countries (LMICs). Although vaccination is the best strategy to prevent rotavirus, obstacles leading to poor vaccine effectiveness undermine its impact in LMICs. This study aimed to identify the optimal rotavirus vaccination strategy for Malawi by modeling vaccine impact and cost-effectiveness, comparing the current two-dose Rotarix vaccine schedule to two alternative vaccine delivery schedules and a next-generation neonatal vaccine (RV3-BB) from 2025-2034. The cost-effectiveness of rotavirus vaccine strategies in Malawi was evaluated from the government and societal perspectives using estimates of moderate-to-severe and non-severe rotavirus cases derived from a transmission dynamic model of rotavirus and published estimates of health-seeking behaviors and costs as inputs. A probabilistic sensitivity analysis was performed to evaluate the robustness of our results to parameter uncertainty. Over a ten-year time horizon, the current two-dose Rotarix strategy is predicted to avert over 1.5 million cases and 90,000 disability-adjusted life-years (DALYs) compared to no vaccination and is cost-effective at willingness-to-pay (WTP) thresholds above $105 per DALY averted from the government perspective. Adding a third dose at 14 weeks could avert an additional 1 million cases and 38,000 DALYs, while switching to the neonatal RV3-BB vaccine could avert 1.1 million cases and 41,000 DALYs compared to the current strategy. Whereas adding a third dose of Rotarix would cost $4.1-4.9 million, switching to the neonatal vaccine is expected to save $3.7 million compared to the current strategy. Considering the neonatal vaccine is not yet available, adding a third dose of Rotarix at 14 weeks of age is cost-effective at WTP thresholds above $138 per DALY averted. The neonatal vaccine offers a more cost-effective alternative to Malawi's current rotavirus vaccine, while adding a third dose to the current strategy also provides substantial benefits.
Collapse
Affiliation(s)
- Catherine Wenger
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
| | - Ernest O. Asare
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
| | - Jiye Kwon
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
| | - Xiao Li
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
- Centre for Health Economics Research and Modelling Infectious Diseases (CHERMID), University of Antwerp, Belgium
| | - Edson Mwinjiwa
- NIHR Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, Merseyside, United Kingdom
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
- Department of Health Systems and Policy, School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Jobiba Chinkhumba
- Department of Health Systems and Policy, School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Khuzwayo C. Jere
- NIHR Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, Merseyside, United Kingdom
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Department of Medical Laboratory Sciences, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Daniel Hungerford
- NIHR Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, Merseyside, United Kingdom
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Nigel A. Cunliffe
- NIHR Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, Merseyside, United Kingdom
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - A. David Paltiel
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
- Department of Health Policy & Management, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
| | - Virginia E. Pitzer
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
3
|
Prunas O, Asare EO, Sajewski E, Li Y, Pithawala Z, Weinberger DM, Warren JL, Armah GE, Cunliffe NA, Iturriza-Gómara M, Lopman BA, Pitzer VE. Global estimates of rotavirus vaccine efficacy and effectiveness: a rapid review and meta-regression analysis. EClinicalMedicine 2025; 81:103122. [PMID: 40115174 PMCID: PMC11925534 DOI: 10.1016/j.eclinm.2025.103122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 03/23/2025] Open
Abstract
Background Rotavirus is the leading cause of diarrhoea worldwide, particularly affecting young children. While national rotavirus immunization programs have reduced rotavirus morbidity and mortality, vaccine performance varies considerably between high-income and low-income settings. Methods We updated a previous systematic review of studies reporting rotavirus vaccine efficacy and vaccine effectiveness against severe rotavirus-associated gastroenteritis (RVGE) by performing a rapid review from July 1, 2020 through October 16, 2024. We included randomized controlled trials reporting vaccine efficacy against severe RVGE and case-control and cohort studies reporting vaccine effectiveness against hospitalization with RVGE in children <5 years old for current internationally licensed vaccines. We developed a meta-regression model for vaccine efficacy and effectiveness using widely available country-specific predictors of rotavirus vaccine performance and simultaneously estimated the relationship between vaccine efficacy and effectiveness. We used the model to predict vaccine efficacy and effectiveness for all countries and assessed its predictive accuracy using a modified leave-one-country-out validation approach. Findings Predicted vaccine efficacy ranged from 69.6% to 94.3% across countries in the Americas, European, and Western Pacific Regions, with a decreased efficacy ranging from 18.6% to 85.3% in the African, South-East Asian, and Eastern Mediterranean regions. Estimates of vaccine effectiveness were generally lower than vaccine efficacy when efficacy was greater than 60%, but effectiveness was predicted to be higher when vaccine efficacy was low. A strong correlation (r = 0.63) was found between the observed and predicted vaccine efficacy and effectiveness, with 98.2% of observed efficacy and effectiveness estimates falling within the 95% prediction intervals. Interpretation Our approach enhances the understanding of global variation in rotavirus vaccine performance and can be used to inform predictions of the potential impact of rotavirus vaccines for countries that have yet to introduce them. Higher-quality data on predictor variables and broader regional representation in vaccine trials are required for more robust vaccine performance estimates. Funding National Institutes of Health/National Institute of Allergy and Infectious Diseases (R01AI112970) and the Bill & Melinda Gates Foundation (INV-17940).
Collapse
Affiliation(s)
- Ottavia Prunas
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Ernest O Asare
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Elizabeth Sajewski
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Yueqi Li
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Zeaan Pithawala
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Daniel M Weinberger
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Joshua L Warren
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - George E Armah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Nigel A Cunliffe
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Miren Iturriza-Gómara
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Benjamin A Lopman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Virginia E Pitzer
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
| |
Collapse
|
4
|
Pun J, Evans C, Chasekwa B, Church JA, Gough E, Mutasa K, Rukobo S, Govha M, Mushayanembwa P, Majo FD, Tavengwa NV, Humphrey JH, Kirkpatrick BD, Kosek M, Ntozini R, Prendergast AJ. Associations Between Histo-blood Group Antigen Status in Mother-Infant Dyads and Infant Oral Rotavirus Vaccine Immunogenicity in Rural Zimbabwe. J Infect Dis 2025; 231:e225-e233. [PMID: 39352457 PMCID: PMC11793023 DOI: 10.1093/infdis/jiae456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/28/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Histo-blood group antigen (HBGA) phenotypes may contribute to poor oral rotavirus vaccine (RVV) immunogenicity, since rotavirus binds intestinal epithelial HBGA glycans, while maternal HBGA status shapes breastmilk composition, which influences the composition of the infant microbiome. We investigated associations between maternal/infant HBGA phenotypes and RVV immunogenicity in rural Zimbabwe. METHODS We undertook salivary FUT2/FUT3 phenotyping in mother-infant pairs. Serum anti-rotavirus immunoglobulin A was measured by enzyme-linked immunosorbent assay. We explored adjusted associations between FUT2/FUT3 status and RVV seroconversion (primary outcome, n = 322) and seropositivity and geometric mean titer (secondary outcomes, n = 776). RESULTS Infants of FUT2- or FUT3-positive women were less likely to seroconvert post-RVV than infants of FUT2- or FUT3-negative women (FUT2 positive [20.1%] vs FUT2 negative [27.5%]: adjusted relative risk [aRR], 0.47; 95% CI, .26-.82; P = .008; FUT3 positive [18.1%] vs FUT3 negative [30.0%]: aRR, 0.45; 95% CI, .25-.78; P = .005). When compared with FUT2-positive infants with FUT2-positive mothers, FUT2-positive infants with FUT2-negative mothers were twice as likely to seroconvert (36.8% vs 21.9%; aRR, 2.12; 95% CI, 1.23-3.63; P = .006). When compared with FUT3-positive infants with FUT3-positive mothers, FUT3-positive infants with FUT3-negative mothers were 3 times as likely to seroconvert (48.3% vs 18.2%; aRR, 2.99; 95% CI, 1.82-4.90; P < .001). CONCLUSIONS Maternal and infant FUT2 and FUT3 status influences infant RVV immunogenicity.
Collapse
Affiliation(s)
- Joshua Pun
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, United Kingdom
| | - Ceri Evans
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, United Kingdom
| | - Bernard Chasekwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - James A Church
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Ethan Gough
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Sandra Rukobo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Margaret Govha
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | | | - Florence D Majo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Naume V Tavengwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Jean H Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Beth D Kirkpatrick
- Vaccine Testing Center, Department of Microbiology and Molecular Genetics, College of Medicine, University of Vermont, Burlington
| | - Margaret Kosek
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville
| | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Andrew J Prendergast
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| |
Collapse
|
5
|
Akinwande KS, Akinduti PA, Arinola O. Rotavirus-specific-IgA and cytokines responses in Ascaris lumbricoides-infected preschool-aged Nigerian children following rotavirus vaccination. J Immunoassay Immunochem 2025; 46:75-88. [PMID: 39533525 DOI: 10.1080/15321819.2024.2426147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Rotavirus diarrhea and Ascaris lumbricoides (Al) infection increase intestinal morbidity and were associated with altered immune responses that compromise the vaccine efficacy in children. The serum level of rotavirus specific IgA (RV-IgA) and cytokine profiles in A. lumbricoides (AI) infected preschool-aged Nigerian children were estimated following oral rotavirus vaccination. Nineteen of the 149 preschool-aged children (aged 6 to 60 months) with Ascaris lumbricoides infection paired with age and sex-matched helminth - free children were administered with oral rotavirus vaccine after intestinal helminth screening using stool sample concentration technique. Separated sera from 3 mL venous blood samples were collected and estimated for cytokines (IFN-γ, TNF-α, IL-4, IL-8 IL-6, IL-10) and RV-IgA before and three weeks after rotavirus vaccination using Enzyme Linked Immunosorbent Assay. IFN-γ, IL-8, IL-4 were significantly lower at post-vaccination in Al-infected children compared with pre-vaccination. Serum IL-10 was significantly higher at post-vaccination in both Al-infected children and helminth-free controls, compared with pre-vaccination levels (p < 0.05). Pre-vaccination IL-8 and IL-6 were significantly higher in Ascaris lumbricoides-infected children, while the post-vaccination IL-8 was significantly higher in Ascaris lumbricoides-infected compared with control. At post-vaccination period, RV-IgA level was lower in Al-infected children and significantly higher in helminth - free control group compared to pre-vaccination RV-IgA level. Ascaris lumbricoides infection contributed to down-regulation of some cytokines and antibody responses to oral rotavirus vaccine.
Collapse
Affiliation(s)
- Kazeem Sanjo Akinwande
- Department of Chemical Pathology and Immunology, Federal Medical Centre, Abeokuta, Nigeria
- Department of Medical Laboratory Science, Chrisland University, Abeokuta, Nigeria
| | | | | |
Collapse
|
6
|
van Dorst MMAR, Pyuza JJ, Nkurunungi G, Kullaya VI, Smits HH, Hogendoorn PCW, Wammes LJ, Everts B, Elliott AM, Jochems SP, Yazdanbakhsh M. Immunological factors linked to geographical variation in vaccine responses. Nat Rev Immunol 2024; 24:250-263. [PMID: 37770632 DOI: 10.1038/s41577-023-00941-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2023] [Indexed: 09/30/2023]
Abstract
Vaccination is one of medicine's greatest achievements; however, its full potential is hampered by considerable variation in efficacy across populations and geographical regions. For example, attenuated malaria vaccines in high-income countries confer almost 100% protection, whereas in low-income regions these same vaccines achieve only 20-50% protection. This trend is also observed for other vaccines, such as bacillus Calmette-Guérin (BCG), rotavirus and yellow fever vaccines, in terms of either immunogenicity or efficacy. Multiple environmental factors affect vaccine responses, including pathogen exposure, microbiota composition and dietary nutrients. However, there has been variable success with interventions that target these individual factors, highlighting the need for a better understanding of their downstream immunological mechanisms to develop new ways of modulating vaccine responses. Here, we review the immunological factors that underlie geographical variation in vaccine responses. Through the identification of causal pathways that link environmental influences to vaccine responsiveness, it might become possible to devise modulatory compounds that can complement vaccines for better outcomes in regions where they are needed most.
Collapse
Affiliation(s)
- Marloes M A R van Dorst
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Jeremia J Pyuza
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
- Department of Pathology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Gyaviira Nkurunungi
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Vesla I Kullaya
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Linda J Wammes
- Department of Medical Microbiology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Alison M Elliott
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Simon P Jochems
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
7
|
Macías-Parra M, Vidal-Vázquez P, Reyna-Figueroa J, Rodríguez-Weber MÁ, Moreno-Macías H, Hernández-Benavides I, Fortes-Gutiérrez S, Richardson VL, Vázquez-Cárdenas P. Immunogenicity of RV1 and RV5 vaccines administered in standard and interchangeable mixed schedules: a randomized, double-blind, non-inferiority clinical trial in Mexican infants. Front Public Health 2024; 12:1356932. [PMID: 38463163 PMCID: PMC10920348 DOI: 10.3389/fpubh.2024.1356932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/06/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction Rotavirus-associated diarrheal diseases significantly burden healthcare systems, particularly affecting infants under five years. Both Rotarix™ (RV1) and RotaTeq™ (RV5) vaccines have been effective but have distinct application schedules and limited interchangeability data. This study aims to provide evidence on the immunogenicity, reactogenicity, and safety of mixed RV1-RV5 schedules compared to their standard counterparts. Methods This randomized, double-blind study evaluated the non-inferiority in terms of immunogenicity of mixed rotavirus vaccine schedules compared to standard RV1 and RV5 schedules in a cohort of 1,498 healthy infants aged 6 to 10 weeks. Participants were randomly assigned to one of seven groups receiving various combinations of RV1, and RV5. Standard RV1 and RV5 schedules served as controls of immunogenicity, reactogenicity, and safety analysis. IgA antibody levels were measured from blood samples collected before the first dose and one month after the third dose. Non-inferiority was concluded if the reduction in seroresponse rate in the mixed schemes, compared to the standard highest responding scheme, did not exceed the non-inferiority margin of -0.10. Reactogenicity traits and adverse events were monitored for 30 days after each vaccination and analyzed on the entire cohort. Results Out of the initial cohort, 1,365 infants completed the study. Immunogenicity analysis included 1,014 infants, considering IgA antibody titers ≥20 U/mL as seropositive. Mixed vaccine schedules demonstrated non-inferiority to standard schedules, with no significant differences in immunogenic response. Safety profiles were comparable across all groups, with no increased incidence of serious adverse events or intussusception. Conclusion The study confirms that mixed rotavirus vaccine schedules are non-inferior to standard RV1 and RV5 regimens in terms of immunogenicity and safety. This finding supports the flexibility of rotavirus vaccination strategies, particularly in contexts of vaccine shortage or logistic constraints. These results contribute to the global effort to optimize rotavirus vaccination programs for broader and more effective pediatric coverage.Clinical trial registration: ClinicalTrials.gov, NCT02193061.
Collapse
Affiliation(s)
| | - Patricia Vidal-Vázquez
- Subdirección de Investigación Biomédica, Hospital General Dr. Manuel Gea González, Mexico City, Mexico
| | - Jesús Reyna-Figueroa
- Unidad de Enfermedades Infecciosas y Epidemiología, Instituto Nacional de Perinatología, Mexico City, Mexico
| | | | | | | | - Sofía Fortes-Gutiérrez
- Subdirección de Investigación Biomédica, Hospital General Dr. Manuel Gea González, Mexico City, Mexico
| | - Vesta Louise Richardson
- Coordinación del Servicio de Guardería para el Desarrollo Integral Infantil, Dirección de Prestaciones Económicas y Sociales, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Paola Vázquez-Cárdenas
- Subdirección de Investigación Biomédica, Hospital General Dr. Manuel Gea González, Mexico City, Mexico
| |
Collapse
|
8
|
Le LKT, Pham TPT, Mai LTP, Nguyen QT, Tran MPN, Ho TH, Pham HH, Le SV, Hoang HN, Lai AT, Huong NT, Nguyen HD, Anh DD, Iijima M, Parashar UD, Trang NV, Tate JE. Intussusception and Other Adverse Event Surveillance after Pilot Introduction of Rotavirus Vaccine in Nam Dinh and Thua Thien Hue Provinces-Vietnam, 2017-2021. Vaccines (Basel) 2024; 12:170. [PMID: 38400153 PMCID: PMC10893515 DOI: 10.3390/vaccines12020170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Rotavin-M1 (POLYVAC) was licensed in Vietnam in 2012. The association of Rotavin-M1 with intussusception, a rare adverse event associated with rotavirus vaccines, and with adverse events following immunization (AEFI) have not been evaluated and monitored under conditions of routine use. From February 2017 to May 2021, we conducted a pilot introduction of Rotavin-M1 into the routine vaccination program in two provinces. Surveillance for intussusception was conducted at six sentinel hospitals. AEFI reports at 30 min and 7 days after vaccination were recorded. Among 443 children <12 months of age admitted for intussusception, most (92.3%) were children ≥ 6 months. Of the 388 children who were age-eligible to receive Rotavin-M1, 116 (29.9%) had received ≥1 dose. No intussusception cases occurred in the 1-21 days after dose 1 and one case occurred on day 21 after dose 2. Among the 45,367 children who received ≥1 dose of Rotavin-M1, 9.5% of children reported at least one AEFI after dose 1 and 7.3% after dose 2. Significantly higher AEFI rates occurred among children given Rotavin-M1 with pentavalent vaccines (Quinvaxem®, ComBE Five®) compared to Rotavin-M1 without pentavalent vaccines. There was no association between intussusception and Rotavin-M1. The vaccine was generally safe when administered alone and when co-administered with other vaccines.
Collapse
Affiliation(s)
- Ly Khanh Thi Le
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Vietnam; (L.K.T.L.); (D.D.A.)
| | - Thao Phuong Thi Pham
- Center for Research and Production of Vaccines and Biologicals, Hanoi 100000, Vietnam; (T.P.T.P.); (N.T.H.)
| | - Le Thi Phuong Mai
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Vietnam; (L.K.T.L.); (D.D.A.)
| | - Quyet Tu Nguyen
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Vietnam; (L.K.T.L.); (D.D.A.)
| | - Mai Phuong Ngoc Tran
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Vietnam; (L.K.T.L.); (D.D.A.)
| | - Thien Huu Ho
- Central Hue Hospital, Thua Thien Hue 530000, Vietnam; (T.H.H.)
| | - Hung Hoang Pham
- Central Hue Hospital, Thua Thien Hue 530000, Vietnam; (T.H.H.)
| | - Sanh Van Le
- Hue Center for Disease Control, Thua Thien Hue 530000, Vietnam
| | | | - Anh Tuan Lai
- Nam Dinh Center for Disease Control, Nam Dinh 420000, Vietnam
| | - Nguyen Thuy Huong
- Center for Research and Production of Vaccines and Biologicals, Hanoi 100000, Vietnam; (T.P.T.P.); (N.T.H.)
| | - Hien Dang Nguyen
- Center for Research and Production of Vaccines and Biologicals, Hanoi 100000, Vietnam; (T.P.T.P.); (N.T.H.)
| | - Dang Duc Anh
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Vietnam; (L.K.T.L.); (D.D.A.)
| | - Makiko Iijima
- World Health Organization, Vietnam Office, Hanoi 100000, Vietnam;
| | - Umesh D. Parashar
- United States Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Nguyen Van Trang
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Vietnam; (L.K.T.L.); (D.D.A.)
| | - Jacqueline E. Tate
- United States Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| |
Collapse
|
9
|
Tate JE, Mwenda JM, Keita AM, Tapsoba TW, Ngendahayo E, Kouamé BD, Samateh AL, Aliabadi N, Sissoko S, Traore Y, Bayisenga J, Sounkere-Soro M, Jagne S, Burke RM, Onwuchekwa U, Ouattara M, Bikoroti JB, N'Zue K, Leshem E, Coulibaly O, Ouedraogo I, Uwimana J, Sow S, Parashar UD. Evaluation of Intussusception Following Pentavalent Rotavirus Vaccine (RotaTeq) Administration in 5 African Countries. Clin Infect Dis 2024; 78:210-216. [PMID: 37596934 PMCID: PMC11497219 DOI: 10.1093/cid/ciad492] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/07/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND A low-level risk of intussusception following rotavirus vaccination has been observed in some settings and may vary by vaccine type. We examined the association between RotaTeq vaccination and intussusception in low-income settings in a pooled analysis from 5 African countries that introduced RotaTeq into their national immunization program. METHODS Active surveillance was conducted at 20 hospitals to identify intussusception cases. A standard case report form was completed for each enrolled child, and vaccination status was determined by review of the child's vaccination card. The pseudo-likelihood adaptation of self-controlled case-series method was used to assess the association between RotaTeq administration and intussusception in the 1-7, 8-21, and 1-21 day periods after each vaccine dose in infants aged 28-245 days. RESULTS Data from 318 infants with confirmed rotavirus vaccination status were analyzed. No clustering of cases occurred in any of the risk windows after any of the vaccine doses. Compared with the background risk of naturally occurring intussusception, no increased risk was observed after dose 1 in the 1-7 day (relative incidence = 2.71; 95% confidence interval [CI] = 0.47-8.03) or the 8-21 day window (relative incidence = 0.77; 95%CI = 0.0-2.69). Similarly, no increased risk of intussusception was observed in any risk window after dose 2 or 3. CONCLUSIONS RotaTeq vaccination was not associated with increased risk of intussusception in this analysis from 5 African countries. This finding mirrors results from similar analyses with other rotavirus vaccines in low-income settings and highlights the need for vaccine-specific and setting-specific risk monitoring.
Collapse
Affiliation(s)
- Jacqueline E Tate
- US Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jason M Mwenda
- World Health Organization Regional Office for Africa, Brazzaville, Congo
| | | | | | | | | | | | - Negar Aliabadi
- US Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Yacouba Traore
- Centre Hospitalier Universitaire Sourou SANOU de Bobo Dioulasso, Bobo Dioulasso, Burkina Faso
| | | | | | - Sheriffo Jagne
- National Public Health Reference Laboratory, Ministry of Health, Banjul, The Gambia
| | - Rachel M Burke
- US Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Ma Ouattara
- World Health Organization Country Office, Ouagadougou, Burkina Faso
| | | | - Kofi N'Zue
- World Health Organization Country Office, Abidjan, Cote d'Ivoire
| | - Eyal Leshem
- US Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Oumar Coulibaly
- Centre Hospitalier Universitaire Gabriel Touré, Bamako, Mali
| | - Issa Ouedraogo
- Ministry of Health, Expanded Program on Immunizations, Ouagadougou, Burkina Faso
| | | | - Samba Sow
- Center for Vaccine Development, Bamako, Mali
| | - Umesh D Parashar
- US Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
10
|
Sadiq A, Khan J. Rotavirus in developing countries: molecular diversity, epidemiological insights, and strategies for effective vaccination. Front Microbiol 2024; 14:1297269. [PMID: 38249482 PMCID: PMC10797100 DOI: 10.3389/fmicb.2023.1297269] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024] Open
Abstract
Rotavirus (RV) causes the loss of numerous children's lives worldwide each year, and this burden is particularly heavy in low- and lower-middle-income countries where access to healthcare is limited. RV epidemiology exhibits a diverse range of genotypes, which can vary in prevalence and impact across different regions. The human genotypes that are most commonly recognized are G1P[8], G2P[4], G3P[8], G4P[8], G8P[8], G9P[8], and G12P[8]. The diversity of rotavirus genotypes presents a challenge in understanding its global distribution and developing effective vaccines. Oral, live-attenuated rotavirus vaccines have undergone evaluation in various contexts, encompassing both low-income and high-income populations, demonstrating their safety and effectiveness. Rotavirus vaccines have been introduced and implemented in over 120 countries, offering an opportunity to assess their effectiveness in diverse settings. However, these vaccines were less effective in areas with more rotavirus-related deaths and lower economic status compared to wealthier regions with fewer rotavirus-related deaths. Despite their lower efficacy, rotavirus vaccines significantly decrease the occurrence of diarrheal diseases and related mortality. They also prove to be cost-effective in regions with a high burden of such diseases. Regularly evaluating the impact, influence, and cost-effectiveness of rotavirus vaccines, especially the newly approved ones for worldwide use, is essential for deciding if these vaccines should be introduced in countries. This is especially important in places with limited resources to determine if a switch to a different vaccine is necessary. Future research in rotavirus epidemiology should focus on a comprehensive understanding of genotype diversity and its implications for vaccine effectiveness. It is crucial to monitor shifts in genotype prevalence and their association with disease severity, especially in high-risk populations. Policymakers should invest in robust surveillance systems to monitor rotavirus genotypes. This data can guide vaccine development and public health interventions. International collaboration and data sharing are vital to understand genotype diversity on a global scale and facilitate the development of more effective vaccines.
Collapse
Affiliation(s)
- Asma Sadiq
- Department of Microbiology, University of Jhang, Jhang, Pakistan
| | - Jadoon Khan
- Department of Allied and Health Sciences, IQRA University, Chak Shahzad Campus, Islamabad, Pakistan
| |
Collapse
|
11
|
Amit LN, John JL, Mori D, Chin AZ, Mosiun AK, Ahmed K. Increase in rotavirus prevalence with the emergence of genotype G9P[8] in replacement of genotype G12P[6] in Sabah, Malaysia. Arch Virol 2023; 168:173. [PMID: 37269384 DOI: 10.1007/s00705-023-05803-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/19/2023] [Indexed: 06/05/2023]
Abstract
Rotaviruses are major causative agents of acute diarrhea in children under 5 years of age in Malaysia. However, a rotavirus vaccine has not been included in the national vaccination program. To date, only two studies have been carried out in the state of Sabah, Malaysia, although children in this state are at risk of diarrheal diseases. Previous studies showed that 16%-17% of cases of diarrhea were caused by rotaviruses and that equine-like G3 rotavirus strains are predominant. Because the prevalence of rotaviruses and their genotype distribution vary over time, this study was conducted at four government healthcare facilities from September 2019 through February 2020. Our study revealed that the proportion of rotavirus diarrhea increased significantly to 37.2% (51/137) after the emergence of the G9P[8] genotype in replacement of the G12P[8] genotype. Although equine-like G3P[8] strains remain the predominant rotaviruses circulating among children, the Sabahan G9P[8] strain belonged to lineage VI and was phylogenetically related to strains from other countries. A comparison of the Sabahan G9 strains with the G9 vaccine strains used in the RotaSiil and Rotavac vaccines revealed several mismatches in neutralizing epitopes, indicating that these vaccines might not be effective in Sabahan children. However, a vaccine trial may be necessary to understand the precise effects of vaccination.
Collapse
Affiliation(s)
- Lia Natasha Amit
- Department of Pathology and Microbiology, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | - Jecelyn Leaslie John
- Borneo Medical and Health Research Centre, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | - Daisuke Mori
- Department of Pathology and Microbiology, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | - Abraham Zefong Chin
- Department of Community and Family Medicine, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | - Andau Konodan Mosiun
- Kunak District Health Office, Ministry of Health Malaysia, Kunak, Sabah, Malaysia
| | - Kamruddin Ahmed
- Department of Pathology and Microbiology, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia.
- Borneo Medical and Health Research Centre, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia.
| |
Collapse
|
12
|
de Oliveira Matos A, Vilela Rodrigues TC, Tiwari S, Dos Santos Dantas PH, Sartori GR, de Carvalho Azevedo VA, Martins Da Silva JH, de Castro Soares S, Silva-Sales M, Sales-Campos H. Immunoinformatics-guided design of a multi-valent vaccine against Rotavirus and Norovirus (ChRNV22). Comput Biol Med 2023; 159:106941. [PMID: 37105111 DOI: 10.1016/j.compbiomed.2023.106941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/17/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023]
Abstract
Rotavirus (RV) and Norovirus (NV) are the main viral etiologic agents of acute gastroenteritis (AG), a serious pediatric condition associated with significant death rates and long-term complications. Anti-RV vaccination has been proved efficient in the reduction of severe AG worldwide, however, the available vaccines are all attenuated and have suboptimal efficiencies in developing countries, where AG leads to substantial disease burden. On the other hand, no NV vaccine has been licensed so far. Therefore, we used immunoinformatics tools to develop a multi-epitope vaccine (ChRNV22) to prevent severe AG by RV and NV. Epitopes were predicted against 17 prevalent genotypes of four structural proteins (NV's VP1, RV's VP4, VP6 and VP7), and then assembled in a chimeric protein, with two small adjuvant sequences (tetanus toxin P2 epitope and a conserved sequence of RV's enterotoxin, NSP4). Simulations of the immune response and interactions with immune receptors indicated the immunogenic properties of ChRNV22, including a Th1-biased response. In silico search for putative host-homologous, allergenic and toxic regions also indicated the vaccine safety. In summary, we developed a multi-epitope vaccine against different NV and RV genotypes that seems promising for the prevention of severe AG, which will be further assessed by in vivo tests.
Collapse
Affiliation(s)
- Amanda de Oliveira Matos
- Laboratory of Mucosal Immunology and Immunoinformatics (LIM), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, 746050-050, Brazil
| | - Thaís Cristina Vilela Rodrigues
- Laboratory of Cellular and Molecular Genetics (LGCM), Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, 31270-901, Brazil
| | - Sandeep Tiwari
- Institute of Biology, Federal University of Bahia (UFBA), Salvador, 40170-115, Brazil; Institute of Health Sciences, Federal University of Bahia (UFBA), Salvador, 40231-300, Brazil
| | - Pedro Henrique Dos Santos Dantas
- Laboratory of Mucosal Immunology and Immunoinformatics (LIM), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, 746050-050, Brazil
| | | | - Vasco Ariston de Carvalho Azevedo
- Laboratory of Cellular and Molecular Genetics (LGCM), Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, 31270-901, Brazil
| | | | - Siomar de Castro Soares
- Department of Immunology, Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM), Uberaba, 38025-180, Brazil
| | - Marcelle Silva-Sales
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, 746050-050, Brazil
| | - Helioswilton Sales-Campos
- Laboratory of Mucosal Immunology and Immunoinformatics (LIM), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, 746050-050, Brazil.
| |
Collapse
|
13
|
Dalton AF, Weber ZA, Allen KS, Stenehjem E, Irving SA, Spark TL, Adams K, Zerbo O, Lazariu V, Dixon BE, Dascomb K, Hartmann E, Kharbanda AB, Ong TC, DeSilva MB, Beaton M, Gaglani M, Patel P, Naleway AL, Kish MNS, Grannis SJ, Grisel N, Sloan-Aagard C, Rao S, Raiyani C, Dickerson M, Bassett E, Fadel WF, Arndorfer J, Nanez J, Barron MA, Vazquez-Benitez G, Liao IC, Griggs EP, Reese SE, Valvi NR, Murthy K, Rowley EAK, Embi PJ, Ball S, Link-Gelles R, Tenforde MW. Relationships Between Social Vulnerability and Coronavirus Disease 2019 Vaccination Coverage and Vaccine Effectiveness. Clin Infect Dis 2023; 76:1615-1625. [PMID: 36611252 PMCID: PMC10949185 DOI: 10.1093/cid/ciad003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/09/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) vaccination coverage remains lower in communities with higher social vulnerability. Factors such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exposure risk and access to healthcare are often correlated with social vulnerability and may therefore contribute to a relationship between vulnerability and observed vaccine effectiveness (VE). Understanding whether these factors impact VE could contribute to our understanding of real-world VE. METHODS We used electronic health record data from 7 health systems to assess vaccination coverage among patients with medically attended COVID-19-like illness. We then used a test-negative design to assess VE for 2- and 3-dose messenger RNA (mRNA) adult (≥18 years) vaccine recipients across Social Vulnerability Index (SVI) quartiles. SVI rankings were determined by geocoding patient addresses to census tracts; rankings were grouped into quartiles for analysis. RESULTS In July 2021, primary series vaccination coverage was higher in the least vulnerable quartile than in the most vulnerable quartile (56% vs 36%, respectively). In February 2022, booster dose coverage among persons who had completed a primary series was higher in the least vulnerable quartile than in the most vulnerable quartile (43% vs 30%). VE among 2-dose and 3-dose recipients during the Delta and Omicron BA.1 periods of predominance was similar across SVI quartiles. CONCLUSIONS COVID-19 vaccination coverage varied substantially by SVI. Differences in VE estimates by SVI were minimal across groups after adjusting for baseline patient factors. However, lower vaccination coverage among more socially vulnerable groups means that the burden of illness is still disproportionately borne by the most socially vulnerable populations.
Collapse
Affiliation(s)
- Alexandra F Dalton
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention COVID-19 Response Team, Atlanta, Georgia, USA
| | | | - Katie S Allen
- Center for Biomedical Informatics, Regenstrief Institute, Indianapolis, Indiana, USA
- Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana, USA
| | - Edward Stenehjem
- Division of Infectious Diseases and Clinical Epidemiology, Intermountain Healthcare, Salt Lake City, Utah, USA
| | - Stephanie A Irving
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon, USA
| | | | - Katherine Adams
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention COVID-19 Response Team, Atlanta, Georgia, USA
| | - Ousseny Zerbo
- Kaiser Permanente Vaccine Study Center, Kaiser Permanente Northern California Division of Research, Oakland, California, USA
| | | | - Brian E Dixon
- Center for Biomedical Informatics, Regenstrief Institute, Indianapolis, Indiana, USA
- Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana, USA
| | - Kristin Dascomb
- Division of Infectious Diseases and Clinical Epidemiology, Intermountain Healthcare, Salt Lake City, Utah, USA
| | - Emily Hartmann
- Paso del Norte Health Information Exchange (PHIX), El Paso, Texas, USA
| | - Anupam B Kharbanda
- Department of Pediatric Emergency Medicine, Children's Minnesota, Minneapolis, Minnesota, USA
| | - Toan C Ong
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Malini B DeSilva
- Division of Research, HealthPartners Institute, Minneapolis, Minnesota, USA
| | - Maura Beaton
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, New York, USA
| | - Manjusha Gaglani
- Baylor Scott & White Health, Temple, Texas, USA
- Texas A&M University College of Medicine, Temple, Texas, USA
| | - Palak Patel
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention COVID-19 Response Team, Atlanta, Georgia, USA
| | - Allison L Naleway
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon, USA
| | | | - Shaun J Grannis
- Center for Biomedical Informatics, Regenstrief Institute, Indianapolis, Indiana, USA
- Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana, USA
| | - Nancy Grisel
- Division of Infectious Diseases and Clinical Epidemiology, Intermountain Healthcare, Salt Lake City, Utah, USA
| | - Chantel Sloan-Aagard
- Paso del Norte Health Information Exchange (PHIX), El Paso, Texas, USA
- Brigham Young University Department of Public Health, Provo, Utah, USA
| | - Suchitra Rao
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | | - Monica Dickerson
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention COVID-19 Response Team, Atlanta, Georgia, USA
| | | | - William F Fadel
- Center for Biomedical Informatics, Regenstrief Institute, Indianapolis, Indiana, USA
- Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana, USA
| | - Julie Arndorfer
- Division of Infectious Diseases and Clinical Epidemiology, Intermountain Healthcare, Salt Lake City, Utah, USA
| | - Juan Nanez
- Paso del Norte Health Information Exchange (PHIX), El Paso, Texas, USA
| | - Michelle A Barron
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | | - I Chia Liao
- Baylor Scott & White Health, Temple, Texas, USA
| | - Eric P Griggs
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention COVID-19 Response Team, Atlanta, Georgia, USA
| | | | - Nimish R Valvi
- Center for Biomedical Informatics, Regenstrief Institute, Indianapolis, Indiana, USA
| | | | | | - Peter J Embi
- Center for Biomedical Informatics, Regenstrief Institute, Indianapolis, Indiana, USA
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Ruth Link-Gelles
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention COVID-19 Response Team, Atlanta, Georgia, USA
| | - Mark W Tenforde
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention COVID-19 Response Team, Atlanta, Georgia, USA
| |
Collapse
|
14
|
Tate JE, Cortese MM, Offit PA, Parashar UD. Rotavirus Vaccines. PLOTKIN'S VACCINES 2023:1005-1024.e11. [DOI: 10.1016/b978-0-323-79058-1.00053-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
15
|
Vetter V, Gardner RC, Debrus S, Benninghoff B, Pereira P. Established and new rotavirus vaccines: a comprehensive review for healthcare professionals. Hum Vaccin Immunother 2022; 18:1870395. [PMID: 33605839 PMCID: PMC8920198 DOI: 10.1080/21645515.2020.1870395] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/02/2020] [Accepted: 12/28/2020] [Indexed: 01/05/2023] Open
Abstract
Robust scientific evidence related to two rotavirus (RV) vaccines available worldwide demonstrates their significant impact on RV disease burden. Improving RV vaccination coverage may result in better RV disease control. To make RV vaccination accessible to all eligible children worldwide and improve vaccine effectiveness in high-mortality settings, research into new RV vaccines continues. Although current and in-development RV vaccines differ in vaccine design, their common goal is the reduction of RV disease risk in children <5 years old for whom disease burden is the most significant. Given the range of RV vaccines available, informed decision-making is essential regarding the choice of vaccine for immunization. This review aims to describe the landscape of current and new RV vaccines, providing context for the assessment of their similarities and differences. As data for new vaccines are limited, future investigations will be required to evaluate their performance/added value in a real-world setting.
Collapse
Affiliation(s)
- Volker Vetter
- Medical Affairs Department, GSK, Wavre, Belgium
- Vaccines R&D – Technical R&D, GSK, Wavre, Belgium
| | - Robert C. Gardner
- Medical Affairs Department, GSK, Wavre, Belgium
- Vaccines R&D – Technical R&D, GSK, Wavre, Belgium
| | - Serge Debrus
- Medical Affairs Department, GSK, Wavre, Belgium
- Vaccines R&D – Technical R&D, GSK, Wavre, Belgium
| | - Bernd Benninghoff
- Medical Affairs Department, GSK, Wavre, Belgium
- Vaccines R&D – Technical R&D, GSK, Wavre, Belgium
| | - Priya Pereira
- Medical Affairs Department, GSK, Wavre, Belgium
- Vaccines R&D – Technical R&D, GSK, Wavre, Belgium
| |
Collapse
|
16
|
Fan Q. A Clinical Nursing Care Study on the Prevalence of Rotavirus Infection and Acute Diarrhea in Vaccinated Chinese Pediatric Population from 2019-2022. Infect Drug Resist 2022; 15:6129-6142. [PMID: 36277240 PMCID: PMC9585908 DOI: 10.2147/idr.s383979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022] Open
Abstract
Purpose To investigate the prevalence of rotavirus infection and acute diarrhea after immunization and further assess the quality of nursing care provided by the nurses to such patients. Methods A total of 432 children aged 3–36 months with acute diarrhea between February 2019 and March 2022 were enrolled, and rotavirus testing was performed within 24 h using a rotavirus enzyme immunoassay kit. Clinical characteristics were evaluated, and regression analysis was performed. Results Eighty vaccinated children (18.5%) were confirmed to have rotavirus infection out of 432 children. The prevalence of rotavirus positivity was the highest at 20–28 months (22 cases, 24.44%) and 11–19 months age group (27 cases, 22.50%). There is a significant association between rotavirus infection and hygiene score (p = 0.009). Based on the association with quality of nursing care, rotavirus infection was association with “appropriate care” (p = 0.001). Conclusion Rotavirus infection was strongly associated with poor hygiene score which may be due to the hygienic nature of the mother and her family. Nursing care assessments revealed a huge gap between nurses and the guardians, which reflects the behavior of Chinese nurses. Thus, an intervention is required by the policymakers for implementing effective strategies of quality nursing for the improvement of the pediatric patients with rotavirus gastroenteritis.
Collapse
Affiliation(s)
- Qiuhua Fan
- Clinical Medical Laboratory Center, Shanxi Children’s Hospital (Shanxi Maternal and Child Health Hospital), Taiyuan, 030000, People’s Republic of China,Correspondence: Qiuhua Fan, Clinical Medical Laboratory Center, Shanxi Children’s Hospital (Shanxi Maternal and Child Health Hospital), Taiyuan, 030000, People’s Republic of China, Tel/Fax +86-13-834209526, Email
| |
Collapse
|
17
|
McAdams D, Estrada M, Holland D, Singh J, Sawant N, Hickey JM, Kumar P, Plikaytis B, Joshi SB, Volkin DB, Sitrin R, Cryz S, White JA. Concordance of in vitro and in vivo measures of non-replicating rotavirus vaccine potency. Vaccine 2022; 40:5069-5078. [PMID: 35871866 PMCID: PMC9405915 DOI: 10.1016/j.vaccine.2022.07.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/16/2022] [Accepted: 07/14/2022] [Indexed: 12/03/2022]
Abstract
Rotavirus infections remain a leading cause of morbidity and mortality among infants residing in low- and middle-income countries. To address the large need for protection from this vaccine-preventable disease we are developing a trivalent subunit rotavirus vaccine which is currently being evaluated in a multinational Phase 3 clinical trial for prevention of serious rotavirus gastroenteritis. Currently, there are no universally accepted in vivo or in vitro models that allow for correlation of field efficacy to an immune response against serious rotavirus gastroenteritis. As a new generation of non-replicating rotavirus vaccines are developed the lack of an established model for evaluating vaccine efficacy becomes a critical issue related to how vaccine potency and stability can be assessed. Our previous publication described the development of an in vitro ELISA to quantify individual vaccine antigens adsorbed to an aluminum hydroxide adjuvant to address the gap in vaccine potency methods for this non-replicating rotavirus vaccine candidate. In the present study, we report on concordance between ELISA readouts and in vivo immunogenicity in a guinea pig model as it relates to vaccine dosing levels and sensitivity to thermal stress. We found correlation between in vitro ELISA values and neutralizing antibody responses engendered after animal immunization. Furthermore, this in vitro assay could be used to demonstrate the effect of thermal stress on vaccine potency, and such results could be correlated with physicochemical analysis of the recombinant protein antigens. This work demonstrates the suitability of the in vitro ELISA to measure vaccine potency and the correlation of these measurements to an immunologic outcome.
Collapse
Affiliation(s)
- David McAdams
- PATH, 2201 Westlake Ave, Seattle, WA 98122, United States
| | - Marcus Estrada
- PATH, 2201 Westlake Ave, Seattle, WA 98122, United States.
| | - David Holland
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, 2030 Becker Drive, Lawrence, KS 66047, United States.
| | - Jasneet Singh
- PATH, 2201 Westlake Ave, Seattle, WA 98122, United States
| | - Nishant Sawant
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, 2030 Becker Drive, Lawrence, KS 66047, United States
| | - John M Hickey
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, 2030 Becker Drive, Lawrence, KS 66047, United States.
| | - Prashant Kumar
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, 2030 Becker Drive, Lawrence, KS 66047, United States.
| | - Brian Plikaytis
- BioStat Consulting, LLC, 10429, Big Canoe, Jasper, GA 30143-5125, United States
| | - Sangeeta B Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, 2030 Becker Drive, Lawrence, KS 66047, United States.
| | - David B Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, 2030 Becker Drive, Lawrence, KS 66047, United States.
| | - Robert Sitrin
- PATH, 2201 Westlake Ave, Seattle, WA 98122, United States.
| | - Stan Cryz
- PATH, 2201 Westlake Ave, Seattle, WA 98122, United States.
| | | |
Collapse
|
18
|
Correa VA, Portilho AI, De Gaspari E. Vaccines, Adjuvants and Key Factors for Mucosal Immune Response. Immunology 2022; 167:124-138. [PMID: 35751397 DOI: 10.1111/imm.13526] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 04/26/2022] [Indexed: 11/29/2022] Open
Abstract
Vaccines are the most effective tool to control infectious diseases, which provoke significant morbidity and mortality. Most vaccines are administered through the parenteral route and can elicit a robust systemic humoral response, but they induce a weak T-cell-mediated immunity and are poor inducers of mucosal protection. Considering that most pathogens enter the body through mucosal surfaces, a vaccine that elicits protection in the first site of contact between the host and the pathogen is promising. However, despite the advantages of mucosal vaccines as good options to confer protection on the mucosal surface, only a few mucosal vaccines are currently approved. In this review, we discuss the impact of vaccine administration in different mucosal surfaces; how appropriate adjuvants enhance the induction of protective mucosal immunity and other factors that can influence the mucosal immune response to vaccines. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Victor Araujo Correa
- Adolfo Lutz Institute, Immunology Center, Av Dr Arnaldo, 355, 11th floor, room 1116, Cerqueira César, São Paulo, SP, Brazil.,São Paulo University, Biomedical Sciences Institute, Graduate Program Interunits in Biotechnology, Av Prof Lineu Prestes, 2415, ICB III, São Paulo, SP, Brazil
| | - Amanda Izeli Portilho
- Adolfo Lutz Institute, Immunology Center, Av Dr Arnaldo, 355, 11th floor, room 1116, Cerqueira César, São Paulo, SP, Brazil.,São Paulo University, Biomedical Sciences Institute, Graduate Program Interunits in Biotechnology, Av Prof Lineu Prestes, 2415, ICB III, São Paulo, SP, Brazil
| | - Elizabeth De Gaspari
- Adolfo Lutz Institute, Immunology Center, Av Dr Arnaldo, 355, 11th floor, room 1116, Cerqueira César, São Paulo, SP, Brazil.,São Paulo University, Biomedical Sciences Institute, Graduate Program Interunits in Biotechnology, Av Prof Lineu Prestes, 2415, ICB III, São Paulo, SP, Brazil
| |
Collapse
|
19
|
Varghese T, Kang G, Steele AD. Understanding Rotavirus Vaccine Efficacy and Effectiveness in Countries with High Child Mortality. Vaccines (Basel) 2022; 10:346. [PMID: 35334978 PMCID: PMC8948967 DOI: 10.3390/vaccines10030346] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/16/2022] [Accepted: 02/19/2022] [Indexed: 02/01/2023] Open
Abstract
Rotavirus claims thousands of lives of children globally every year with a disproportionately high burden in low- and lower-middle income countries where access to health care is limited. Oral, live-attenuated rotavirus vaccines have been evaluated in multiple settings in both low- and high-income populations and have been shown to be safe and efficacious. However, the vaccine efficacy observed in low-income settings with high rotavirus and diarrheal mortality was significantly lower than that seen in high-income populations where rotavirus mortality is less common. Rotavirus vaccines have been introduced and rolled out in more than 112 countries, providing the opportunity to assess effectiveness of the vaccines in these different settings. We provide an overview of the efficacy, effectiveness, and impact of rotavirus vaccines, focusing on high-mortality settings and identify the knowledge gaps for future research. Despite lower efficacy, rotavirus vaccines substantially reduce diarrheal disease and mortality and are cost-effective in countries with high burden. Continued evaluation of the effectiveness, impact, and cost-benefit of rotavirus vaccines, especially the new candidates that have been recently approved for global use, is a key factor for new vaccine introductions in countries, or for a switch of vaccine product in countries with limited resources.
Collapse
Affiliation(s)
- Tintu Varghese
- The Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College, Vellore 632004, India; (T.V.); (G.K.)
| | - Gagandeep Kang
- The Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College, Vellore 632004, India; (T.V.); (G.K.)
| | - Andrew Duncan Steele
- Enteric and Diarrheal Disease, Bill & Melinda Gates Foundation, Seattle, WA 98102, USA
| |
Collapse
|
20
|
Afchangi A, Jalilvand S, Arashkia A, Latifi T, Farahmand M, Abolghasem Shirazi MM, Mousavi Nasab SD, Marashi SM, Roohvand F, Shoja Z. Co-administration of rotavirus nanospheres VP6 and NSP4 proteins enhanced the anti-NSP4 humoral responses in immunized mice. Microb Pathog 2022; 163:105405. [PMID: 35045328 DOI: 10.1016/j.micpath.2022.105405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/28/2021] [Accepted: 01/12/2022] [Indexed: 11/29/2022]
Abstract
Inconveniences associated with the efficacy and safety of the World Health Organization (WHO) approved/prequalified live attenuated rotavirus (RV) vaccines, sounded for finding alternative non-replicating modals and proper RV antigens (Ags). Herein, we report the development of a RV candidate vaccine based on the combination of RV VP6 nanospheres (S) and NSP4112-175 proteins (VP6S + NSP4). Self-assembled VP6S protein was produced in insect cells. Analyses by western blotting and transmission electron microscopy (TEM) indicated expression of VP6 trimer structures with sizes of ≥140 kDa and presence of VP6S. Four group of mice were immunized (2-dose formulation) intra-peritoneally (IP) by either¨VP6S + NSP4¨ or each protein alone (VP6S or NSP4112-175) emulsified in aluminium hydroxide or control. Results indicated that VP6S + NSP4 formulation induced significant anti-VP6 IgG (P < 0.001) and IgA (P < 0.05) as well as anti-NSP4 IgG (P < 0.001) and enhancement of protective immunity. Analyses of anti-VP6S and anti-NSP4 IgG subclass (IgG1 and IgG2a) showed IgG1/IgG2a ≥6 and IgG1/IgG2a ≥3 ratios, respectively indicating Th2 polarization of immune responses. The combination of VP6S + NSP4 proteins emulsified in aluminum hydroxide adjuvant might present a dual universal, efficient and cost-effective candidate vaccine against RV infection.
Collapse
Affiliation(s)
- Atefeh Afchangi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Jalilvand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Arash Arashkia
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Tayebeh Latifi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Farahmand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Seyed Dawood Mousavi Nasab
- Department of Research and Development, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Sayed Mahdi Marashi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzin Roohvand
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Zabihollah Shoja
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
21
|
Sadiq A, Bostan N, Aziz A. Effect of rotavirus genetic diversity on vaccine impact. Rev Med Virol 2022; 32:e2259. [PMID: 34997676 DOI: 10.1002/rmv.2259] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 05/05/2021] [Indexed: 11/07/2022]
Abstract
Group A rotaviruses (RVAs) are the leading cause of gastroenteritis, causing 0.2 million deaths and several million hospitalisations globally each year. Four rotavirus vaccines (RotarixTM , RotaTeqTM , Rotavac® and ROTASIIL® ) have been pre-qualified by the World Health Organization (WHO), but the two newly pre-qualified vaccines (Rotavac® and ROTASIIL® ) are currently only in use in Palestine and India, respectively. In 2009, WHO strongly proposed that rotavirus vaccines be included in the routine vaccination schedule of all countries around the world. By the end of 2019, a total of 108 countries had administered rotavirus vaccines, and 10 countries have currently been approved by Gavi for the introduction of rotavirus vaccine in the near future. With 39% of global coverage, rotavirus vaccines have had a substantial effect on diarrhoeal morbidity and mortality in different geographical areas, although efficacy appears to be higher in high income settings. Due to the segmented RNA genome, the pattern of RVA genotypes in the human population is evolving through interspecies transmission and/or reassortment events for which the vaccine might be less effective in the future. However, despite the relative increase in some particular genotypes after rotavirus vaccine use, the overall efficacy of rotavirus mass vaccination worldwide has not been affected. Some of the challenges to improve the effect of current rotavirus vaccines can be solved in the future by new rotavirus vaccines and by vaccines currently in progress.
Collapse
Affiliation(s)
- Asma Sadiq
- Department of Biosciences, Molecular Virology Laboratory, COMSATS University, Islamabad, Pakistan
| | - Nazish Bostan
- Department of Biosciences, Molecular Virology Laboratory, COMSATS University, Islamabad, Pakistan
| | - Aamir Aziz
- Sarhad University of Science and Information Technology, Institute of Biological Sciences, Sarhad University, Peshawar, Pakistan
| |
Collapse
|
22
|
Church JA, Rukobo S, Govha M, Gough EK, Chasekwa B, Lee B, Carmolli MP, Panic G, Giallourou N, Ntozini R, Mutasa K, McNeal MM, Majo FD, Tavengwa NV, Swann JR, Moulton LH, Kirkpatrick BD, Humphrey JH, Prendergast AJ. Associations between biomarkers of environmental enteric dysfunction and oral rotavirus vaccine immunogenicity in rural Zimbabwean infants. EClinicalMedicine 2021; 41:101173. [PMID: 34825149 PMCID: PMC8605235 DOI: 10.1016/j.eclinm.2021.101173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 10/01/2021] [Accepted: 10/11/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Oral rotavirus vaccines (RVV) are poorly immunogenic in low-income countries. Environmental enteric dysfunction (EED) resulting from poor water, sanitation and hygiene (WASH) may contribute. We therefore tested associations between EED and RVV immunogenicity, and evaluated the effect of improved WASH on EED. METHODS We measured nine biomarkers of EED among Zimbabwean infants born to mothers enrolled in a cluster-randomised 2 × 2 factorial trial of improved WASH and improved feeding between November 2012 and March 2015 (NCT01824940). We used multivariable regression to determine associations between EED biomarkers and RVV seroconversion, seropositivity and geometric mean titer. Log-binomial regression was used to evaluate the effect of improved WASH on EED. FINDINGS Among 303 infants with EED biomarkers and immunogenicity data, plasma intestinal fatty-acid binding protein and stool myeloperoxidase were positively associated with RVV seroconversion; adjusted RR 1.63 (95%CI 1.04, 2.57) and 1.29 (95%CI 1.01, 1.65), respectively. There were no other associations between RVV immunogenicity and either individual biomarkers or EED domains (intestinal permeability, intestinal damage, intestinal inflammation and microbial translocation). EED biomarkers did not differ between randomised WASH and non-WASH groups. INTERPRETATION We found no evidence that EED was associated with poor RVV immunogenicity. Contrary to our hypothesis, there was weak evidence that EED was associated with increased seroconversion. EED biomarkers were not affected by a package of household-level WASH interventions.
Collapse
Affiliation(s)
- James A Church
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
- Corresponding authors at: Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK.
| | - Sandra Rukobo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Margaret Govha
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Ethan K Gough
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Bernard Chasekwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Benjamin Lee
- Departments of Pediatrics, Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Marya P Carmolli
- Departments of Microbiology and Molecular Genetics, Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Gordana Panic
- Faculty of Medicine, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Natasa Giallourou
- Faculty of Medicine, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Monica M McNeal
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Florence D. Majo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Naume V. Tavengwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Jonathan R. Swann
- Faculty of Medicine, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Lawrence H Moulton
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Beth D Kirkpatrick
- Departments of Microbiology and Molecular Genetics, Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Jean H Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Corresponding authors at: Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK.
| |
Collapse
|
23
|
Kurokawa N, Robinson MK, Bernard C, Kawaguchi Y, Koujin Y, Koen A, Madhi S, Polasek TM, McNeal M, Dargis M, Couture MMJ, Trépanier S, Forrest BD, Tsutsui N. Safety and immunogenicity of a plant-derived rotavirus-like particle vaccine in adults, toddlers and infants. Vaccine 2021; 39:5513-5523. [PMID: 34454786 DOI: 10.1016/j.vaccine.2021.08.052] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/06/2021] [Accepted: 08/12/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND This study is the first clinical trial for a parenteral non-replicating rotavirus vaccine developed using virus-like particle (VLP) technology. METHODS This open-labeled, randomized, placebo-controlled trial was conducted in two parts: Part A (a first-in-human study in Australian adults) and Part B (ascending dose and descending age in South African adults, toddlers and infants). In Part A, two cohorts of 10 adults were assigned to receive a single intramuscular injection of 1 of 2 escalating dose levels of the rotavirus VLP (Ro-VLP) vaccine (7 μg or 21 μg) or placebo. In Part B, one cohort of 10 adults was assigned to receive a single injection of the Ro-VLP vaccine (21 μg) or placebo, two cohorts of 10 toddlers were assigned to receive 2 injections of 1 of 2 escalating dose levels of the Ro-VLP vaccine (7 μg or 21 μg) or placebo 28 days apart, and three cohorts of 20 infants were assigned to receive 3 injections of 1 of 3 escalating dose levels of the Ro-VLP vaccine (2.5 μg, 7 μg or 21 μg) or placebo or 2 doses of oral Rotarix 28 days apart. Safety, reactogenicity and immunogenicity were assessed. RESULTS There were no safety or tolerability concerns after administration of the Ro-VLP vaccine. The Ro-VLP vaccine induced an anti-G1P[8] IgG response in infants 4 weeks after the second and third doses. Neutralizing antibody responses against homologous G1P[8] rotavirus were higher in all Ro-VLP infant groups than in the placebo group 4 weeks after the third dose. No heterotypic immunity was elicited by the Ro-VLP vaccine. CONCLUSIONS The Ro-VLP vaccine was well tolerated and induced a homotypic immune response in infants, suggesting that this technology platform is a favorable approach for a parenteral non-replicating rotavirus vaccine. CLINICAL TRIAL REGISTRATION NCT03507738.
Collapse
Affiliation(s)
- Natsuki Kurokawa
- Mitsubishi Tanabe Pharma Corporation, 17-10, Nihonbashi-Koamicho, Chuo-ku, Tokyo 103-8405, Japan.
| | | | - Catherine Bernard
- International Regulatory Affairs Services, Inc., 10626 Wagon Box Way, Highlands Ranch, CO 80130, USA
| | - Yutaka Kawaguchi
- Mitsubishi Tanabe Pharma Corporation, 17-10, Nihonbashi-Koamicho, Chuo-ku, Tokyo 103-8405, Japan
| | - Yoshito Koujin
- Mitsubishi Tanabe Pharma Corporation, 17-10, Nihonbashi-Koamicho, Chuo-ku, Tokyo 103-8405, Japan
| | - Anthonet Koen
- Respiratory and Meningeal Pathogens Research Unit, Chris Hani Baragwanath Hospital, Berstham Chris Hani Road, Soweto 2013, South Africa
| | - Shabir Madhi
- Respiratory and Meningeal Pathogens Research Unit, Chris Hani Baragwanath Hospital, Berstham Chris Hani Road, Soweto 2013, South Africa
| | - Thomas M Polasek
- Department of Clinical Pharmacology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia
| | - Monica McNeal
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| | - Michèle Dargis
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada
| | - Manon M-J Couture
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada
| | - Sonia Trépanier
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada
| | - Bruce D Forrest
- Cognoscenti Bioscience, LLC., PO Box 444, Nyack, NY 10960, USA
| | - Naohisa Tsutsui
- Mitsubishi Tanabe Pharma Corporation, 17-10, Nihonbashi-Koamicho, Chuo-ku, Tokyo 103-8405, Japan
| |
Collapse
|
24
|
Robertson RC, Church JA, Edens TJ, Mutasa K, Min Geum H, Baharmand I, Gill SK, Ntozini R, Chasekwa B, Carr L, Majo FD, Kirkpatrick BD, Lee B, Moulton LH, Humphrey JH, Prendergast AJ, Manges AR. The fecal microbiome and rotavirus vaccine immunogenicity in rural Zimbabwean infants. Vaccine 2021; 39:5391-5400. [PMID: 34393020 PMCID: PMC8423000 DOI: 10.1016/j.vaccine.2021.07.076] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 07/15/2021] [Accepted: 07/27/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Oral rotavirus vaccine (RVV) immunogenicity is considerably lower in low- versus high-income populations; however, the mechanisms underlying this remain unclear. Previous evidence suggests that the gut microbiota may contribute to differences in oral vaccine efficacy. METHODS We performed whole metagenome shotgun sequencing on stool samples and measured anti-rotavirus immunoglobulin A in plasma samples from a subset of infants enrolled in a cluster randomized 2 × 2 factorial trial of improved water, sanitation and hygiene and infant feeding in rural Zimbabwe (SHINE trial: NCT01824940). We examined taxonomic microbiome composition and functional metagenome features using random forest models, differential abundance testing and regression analyses to explored associations with RVV immunogenicity. RESULTS Among 158 infants with stool samples and anti-rotavirus IgA titres, 34 were RVV seroconverters. The median age at stool collection was 43 days (IQR: 35-68), corresponding to a median of 4 days before the first RVV dose. The infant microbiome was dominated by Bifidobacterium longum. The gut microbiome differed significantly between early (≤42 days) and later samples (>42 days) however, we observed no meaningful differences in alpha diversity, beta diversity, species composition or functional metagenomic features by RVV seroconversion status. Bacteroides thetaiotaomicron was the only species associated with anti-rotavirus IgA titre. Random forest models poorly classified seroconversion status by both composition and functional microbiome variables. CONCLUSIONS RVV immunogenicity is low in this rural Zimbabwean setting, however it was not associated with the composition or function of the early-life gut microbiome in this study. Further research is warranted to examine the mechanisms of poor oral RVV efficacy in low-income countries.
Collapse
Affiliation(s)
- Ruairi C Robertson
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK.
| | - James A Church
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Thaddeus J Edens
- Devil's Staircase Consulting, West Vancouver, British Columbia, Canada
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Hyun Min Geum
- British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
| | - Iman Baharmand
- British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
| | - Sandeep K Gill
- British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada; School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Bernard Chasekwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Lynnea Carr
- British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Canada
| | - Florence D Majo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Beth D Kirkpatrick
- Vaccine Testing Center, College of Medicine, University of Vermont, Burlington, VT, USA
| | - Benjamin Lee
- Vaccine Testing Center, College of Medicine, University of Vermont, Burlington, VT, USA
| | - Lawrence H Moulton
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jean H Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew J Prendergast
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Amee R Manges
- British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada; School of Population and Public Health, University of British Columbia, Vancouver, Canada
| |
Collapse
|
25
|
Toor J, Echeverria-Londono S, Li X, Abbas K, Carter ED, Clapham HE, Clark A, de Villiers MJ, Eilertson K, Ferrari M, Gamkrelidze I, Hallett TB, Hinsley WR, Hogan D, Huber JH, Jackson ML, Jean K, Jit M, Karachaliou A, Klepac P, Kraay A, Lessler J, Li X, Lopman BA, Mengistu T, Metcalf CJE, Moore SM, Nayagam S, Papadopoulos T, Perkins TA, Portnoy A, Razavi H, Razavi-Shearer D, Resch S, Sanderson C, Sweet S, Tam Y, Tanvir H, Tran Minh Q, Trotter CL, Truelove SA, Vynnycky E, Walker N, Winter A, Woodruff K, Ferguson NM, Gaythorpe KAM. Lives saved with vaccination for 10 pathogens across 112 countries in a pre-COVID-19 world. eLife 2021; 10:e67635. [PMID: 34253291 PMCID: PMC8277373 DOI: 10.7554/elife.67635] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Background Vaccination is one of the most effective public health interventions. We investigate the impact of vaccination activities for Haemophilus influenzae type b, hepatitis B, human papillomavirus, Japanese encephalitis, measles, Neisseria meningitidis serogroup A, rotavirus, rubella, Streptococcus pneumoniae, and yellow fever over the years 2000-2030 across 112 countries. Methods Twenty-one mathematical models estimated disease burden using standardised demographic and immunisation data. Impact was attributed to the year of vaccination through vaccine-activity-stratified impact ratios. Results We estimate 97 (95%CrI[80, 120]) million deaths would be averted due to vaccination activities over 2000-2030, with 50 (95%CrI[41, 62]) million deaths averted by activities between 2000 and 2019. For children under-5 born between 2000 and 2030, we estimate 52 (95%CrI[41, 69]) million more deaths would occur over their lifetimes without vaccination against these diseases. Conclusions This study represents the largest assessment of vaccine impact before COVID-19-related disruptions and provides motivation for sustaining and improving global vaccination coverage in the future. Funding VIMC is jointly funded by Gavi, the Vaccine Alliance, and the Bill and Melinda Gates Foundation (BMGF) (BMGF grant number: OPP1157270 / INV-009125). Funding from Gavi is channelled via VIMC to the Consortium's modelling groups (VIMC-funded institutions represented in this paper: Imperial College London, London School of Hygiene and Tropical Medicine, Oxford University Clinical Research Unit, Public Health England, Johns Hopkins University, The Pennsylvania State University, Center for Disease Analysis Foundation, Kaiser Permanente Washington, University of Cambridge, University of Notre Dame, Harvard University, Conservatoire National des Arts et Métiers, Emory University, National University of Singapore). Funding from BMGF was used for salaries of the Consortium secretariat (authors represented here: TBH, MJ, XL, SE-L, JT, KW, NMF, KAMG); and channelled via VIMC for travel and subsistence costs of all Consortium members (all authors). We also acknowledge funding from the UK Medical Research Council and Department for International Development, which supported aspects of VIMC's work (MRC grant number: MR/R015600/1).JHH acknowledges funding from National Science Foundation Graduate Research Fellowship; Richard and Peggy Notebaert Premier Fellowship from the University of Notre Dame. BAL acknowledges funding from NIH/NIGMS (grant number R01 GM124280) and NIH/NIAID (grant number R01 AI112970). The Lives Saved Tool (LiST) receives funding support from the Bill and Melinda Gates Foundation.This paper was compiled by all coauthors, including two coauthors from Gavi. Other funders had no role in study design, data collection, data analysis, data interpretation, or writing of the report. All authors had full access to all the data in the study and had final responsibility for the decision to submit for publication.
Collapse
Affiliation(s)
- Jaspreet Toor
- MRC Centre for Global Infectious Disease Analysis; and the Abdul Latif Jameel Institute for Disease and Emergency Analytics (J-IDEA), School of Public Health, Imperial College LondonLondonUnited Kingdom
| | - Susy Echeverria-Londono
- MRC Centre for Global Infectious Disease Analysis; and the Abdul Latif Jameel Institute for Disease and Emergency Analytics (J-IDEA), School of Public Health, Imperial College LondonLondonUnited Kingdom
| | - Xiang Li
- MRC Centre for Global Infectious Disease Analysis; and the Abdul Latif Jameel Institute for Disease and Emergency Analytics (J-IDEA), School of Public Health, Imperial College LondonLondonUnited Kingdom
| | - Kaja Abbas
- London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Emily D Carter
- Bloomberg School of Public Health, Johns Hopkins UniversityBaltimoreUnited States
| | - Hannah E Clapham
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore; Oxford University Clinical Research Unit, Vietnam; Nuffield Department of Medicine, Oxford UniversityOxfordUnited Kingdom
| | - Andrew Clark
- London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Margaret J de Villiers
- MRC Centre for Global Infectious Disease Analysis; and the Abdul Latif Jameel Institute for Disease and Emergency Analytics (J-IDEA), School of Public Health, Imperial College LondonLondonUnited Kingdom
| | | | | | | | - Timothy B Hallett
- MRC Centre for Global Infectious Disease Analysis; and the Abdul Latif Jameel Institute for Disease and Emergency Analytics (J-IDEA), School of Public Health, Imperial College LondonLondonUnited Kingdom
| | - Wes R Hinsley
- MRC Centre for Global Infectious Disease Analysis; and the Abdul Latif Jameel Institute for Disease and Emergency Analytics (J-IDEA), School of Public Health, Imperial College LondonLondonUnited Kingdom
| | | | - John H Huber
- Department of Biological Sciences, University of Notre DameNotre DameUnited States
| | | | - Kevin Jean
- MRC Centre for Global Infectious Disease Analysis; and the Abdul Latif Jameel Institute for Disease and Emergency Analytics (J-IDEA), School of Public Health, Imperial College LondonLondonUnited Kingdom
- Laboratoire MESuRS and Unite PACRI, Institut Pasteur, Conservatoire National des Arts et MetiersParisFrance
| | - Mark Jit
- London School of Hygiene and Tropical MedicineLondonUnited Kingdom
- University of Hong Kong, Hong Kong Special Administrative RegionHong KongChina
| | | | - Petra Klepac
- London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Alicia Kraay
- Rollins School of Public Health, Emory UniversityAtlantaUnited States
| | - Justin Lessler
- Bloomberg School of Public Health, Johns Hopkins UniversityBaltimoreUnited States
| | - Xi Li
- IndependentAtlantaUnited States
| | - Benjamin A Lopman
- Rollins School of Public Health, Emory UniversityAtlantaUnited States
| | | | | | - Sean M Moore
- Department of Biological Sciences, University of Notre DameNotre DameUnited States
| | - Shevanthi Nayagam
- MRC Centre for Global Infectious Disease Analysis; and the Abdul Latif Jameel Institute for Disease and Emergency Analytics (J-IDEA), School of Public Health, Imperial College LondonLondonUnited Kingdom
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Imperial College LondonLondonUnited Kingdom
| | - Timos Papadopoulos
- Public Health EnglandLondonUnited Kingdom
- University of SouthamptonSouthamptonUnited Kingdom
| | - T Alex Perkins
- Department of Biological Sciences, University of Notre DameNotre DameUnited States
| | - Allison Portnoy
- Center for Health Decision Science, Harvard T H Chan School of Public Health, Harvard UniversityCambridgeUnited States
| | - Homie Razavi
- Center for Disease Analysis FoundationLafayetteUnited States
| | | | - Stephen Resch
- Center for Health Decision Science, Harvard T H Chan School of Public Health, Harvard UniversityCambridgeUnited States
| | - Colin Sanderson
- London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Steven Sweet
- Center for Health Decision Science, Harvard T H Chan School of Public Health, Harvard UniversityCambridgeUnited States
| | - Yvonne Tam
- Bloomberg School of Public Health, Johns Hopkins UniversityBaltimoreUnited States
| | - Hira Tanvir
- London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Quan Tran Minh
- Department of Biological Sciences, University of Notre DameNotre DameUnited States
| | | | - Shaun A Truelove
- Bloomberg School of Public Health, Johns Hopkins UniversityBaltimoreUnited States
| | | | - Neff Walker
- Bloomberg School of Public Health, Johns Hopkins UniversityBaltimoreUnited States
| | - Amy Winter
- Bloomberg School of Public Health, Johns Hopkins UniversityBaltimoreUnited States
| | - Kim Woodruff
- MRC Centre for Global Infectious Disease Analysis; and the Abdul Latif Jameel Institute for Disease and Emergency Analytics (J-IDEA), School of Public Health, Imperial College LondonLondonUnited Kingdom
| | - Neil M Ferguson
- MRC Centre for Global Infectious Disease Analysis; and the Abdul Latif Jameel Institute for Disease and Emergency Analytics (J-IDEA), School of Public Health, Imperial College LondonLondonUnited Kingdom
| | - Katy AM Gaythorpe
- MRC Centre for Global Infectious Disease Analysis; and the Abdul Latif Jameel Institute for Disease and Emergency Analytics (J-IDEA), School of Public Health, Imperial College LondonLondonUnited Kingdom
| |
Collapse
|
26
|
Varre JV. Vaccines are not one size fits all, just like medications: rotavirus vaccine study. Clin Exp Vaccine Res 2021; 10:148-150. [PMID: 34222127 PMCID: PMC8217582 DOI: 10.7774/cevr.2021.10.2.148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 12/31/2020] [Accepted: 05/06/2021] [Indexed: 11/23/2022] Open
Abstract
The current global coronavirus disease 2019 pandemic has shown us once again how important vaccination is in controlling and preventing the spread of deadly diseases. Vaccinations are one of the most tried and tested public health measures aimed at the prevention and eventual eradication of various diseases. Many debilitating diseases like polio have been eradicated in countries like India due to effective vaccination strategies. Just like with any other public health initiative, there do exist various challenges for vaccination. Efficacy and correlate of protection studies are crucial in determining which vaccine works best. The rotavirus vaccine (ROTAVAC; Bharat Biotech International Ltd., Hyderabad, India) is one such example where efficacy seen in one geographical and ethnic population is not replicated elsewhere. This has prompted various researchers and pharmaceutical companies to think about customizing vaccines to the individual needs of a particular geographic and ethnic group. In this brief communication, we look at the rotavirus vaccination story and see how it laid down the idea for customized vaccination development and what the future of vaccine development looks like.
Collapse
Affiliation(s)
- Joseph Vinod Varre
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
27
|
Bullington BW, Klemperer K, Mages K, Chalem A, Mazigo HD, Changalucha J, Kapiga S, Wright PF, Yazdanbakhsh MM, Downs JA. Effects of schistosomes on host anti-viral immune response and the acquisition, virulence, and prevention of viral infections: A systematic review. PLoS Pathog 2021; 17:e1009555. [PMID: 34015063 PMCID: PMC8172021 DOI: 10.1371/journal.ppat.1009555] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/02/2021] [Accepted: 04/13/2021] [Indexed: 11/18/2022] Open
Abstract
Although a growing number of studies suggest interactions between Schistosoma parasites and viral infections, the effects of schistosome infections on the host response to viruses have not been evaluated comprehensively. In this systematic review, we investigated how schistosomes impact incidence, virulence, and prevention of viral infections in humans and animals. We also evaluated immune effects of schistosomes in those coinfected with viruses. We screened 4,730 studies and included 103. Schistosomes may increase susceptibility to some viruses, including HIV and Kaposi’s sarcoma-associated herpesvirus, and virulence of hepatitis B and C viruses. In contrast, schistosome infection may be protective in chronic HIV, Human T-cell Lymphotropic Virus-Type 1, and respiratory viruses, though further research is needed. Schistosome infections were consistently reported to impair immune responses to hepatitis B and possibly measles vaccines. Understanding the interplay between schistosomes and viruses has ramifications for anti-viral vaccination strategies and global control of viral infections. Many studies have described the effects of parasitic Schistosoma worm infections on the way that humans and animals respond to a variety of viral infections. Our goal was to evaluate, in a systematic manner, how having a schistosome parasitic infection affects a host’s susceptibility to viral infections, the clinical disease course of viral infections, and prevention of viral infections by vaccines. We also assessed the effects of schistosome infection on the host immune response to viruses. We screened 4,730 studies for potential relevance and included 103 of them in this review. Overall, our analysis showed that schistosome infection impairs the host response to many viruses. This includes increasing host susceptibility to HIV and possibly Kaposi’s sarcoma-associated herpesvirus, worsening the severity of clinical disease in hepatitis B and C infections, and decreasing immune responses to vaccines for hepatitis B and possibly measles. The studies that we analyzed also suggested that schistosome infection may protect the host against poor clinical outcomes from some viral infections including Human T-cell Lymphotropic Virus-Type 1, respiratory viruses, and chronic HIV. We discuss how these findings might be interpreted, and the additional research needed, in order to improve anti-viral vaccination strategies and control of viral infections globally.
Collapse
Affiliation(s)
- Brooke W. Bullington
- Center for Global Health, Weill Cornell Medicine, New York, NY, United States of America
- * E-mail:
| | | | - Keith Mages
- Samuel J. Wood Library Weill Cornell Medicine, New York, NY, United States of America
| | - Andrea Chalem
- Center for Global Health, Weill Cornell Medicine, New York, NY, United States of America
| | - Humphrey D. Mazigo
- Mwanza Intervention Trials Unit, National Institute for Medical Research Mwanza, Tanzania
| | - John Changalucha
- Mwanza Intervention Trials Unit, National Institute for Medical Research Mwanza, Tanzania
| | - Saidi Kapiga
- Mwanza Intervention Trials Unit, National Institute for Medical Research Mwanza, Tanzania
- Department of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Peter F. Wright
- Department of Pediatrics, Dartmouth Geisel School of Medicine, Hanover, New Hampshire, United States of America
| | | | - Jennifer A. Downs
- Center for Global Health, Weill Cornell Medicine, New York, NY, United States of America
| |
Collapse
|
28
|
Salmon DA, Lambert PH, Nohynek HM, Gee J, Parashar UD, Tate JE, Wilder-Smith A, Hartigan-Go KY, Smith PG, Zuber PLF. Novel vaccine safety issues and areas that would benefit from further research. BMJ Glob Health 2021; 6:e003814. [PMID: 34011502 PMCID: PMC8137224 DOI: 10.1136/bmjgh-2020-003814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/01/2020] [Accepted: 01/06/2021] [Indexed: 12/20/2022] Open
Abstract
Vaccine licensure requires a very high safety standard and vaccines routinely used are very safe. Vaccine safety monitoring prelicensure and postlicensure enables continual assessment to ensure the benefits outweigh the risks and, when safety problems arise, they are quickly identified, characterised and further problems prevented when possible. We review five vaccine safety case studies: (1) dengue vaccine and enhanced dengue disease, (2) pandemic influenza vaccine and narcolepsy, (3) rotavirus vaccine and intussusception, (4) human papillomavirus vaccine and postural orthostatic tachycardia syndrome and complex regional pain syndrome, and (5) RTS,S/adjuvant system 01 malaria vaccine and meningitis, cerebral malaria, female mortality and rebound severe malaria. These case studies were selected because they are recent and varied in the vaccine safety challenges they elucidate. Bringing these case studies together, we develop lessons learned that can be useful for addressing some of the potential safety issues that will inevitably arise with new vaccines.
Collapse
Affiliation(s)
- Daniel A Salmon
- Global Disease Epidemiology and Control, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | | - Hanna M Nohynek
- Infectious Disease Control and Vaccinations Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Uusimaa, Finland
| | - Julianne Gee
- Division of Healthcare Quality Promotion, National Center of Emerging and Zoonotic Infectious Diseases, CDC, Atlanta, Georgia, USA
| | - Umesh D Parashar
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, CDC, Atlanta, Georgia, USA
| | - Jacqueline E Tate
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, CDC, Atlanta, Georgia, USA
| | | | | | - Peter G Smith
- MRC Tropical Epidemiology Group, London School of Hygiene & Tropical Medicine, London, London, UK
| | - Patrick Louis F Zuber
- Essential Medicines and Health Products, Organisation Mondiale de la Sante, Geneve, Switzerland
| |
Collapse
|
29
|
Cárcamo-Calvo R, Muñoz C, Buesa J, Rodríguez-Díaz J, Gozalbo-Rovira R. The Rotavirus Vaccine Landscape, an Update. Pathogens 2021; 10:520. [PMID: 33925924 PMCID: PMC8145439 DOI: 10.3390/pathogens10050520] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 11/17/2022] Open
Abstract
Rotavirus is the leading cause of severe acute childhood gastroenteritis, responsible for more than 128,500 deaths per year, mainly in low-income countries. Although the mortality rate has dropped significantly since the introduction of the first vaccines around 2006, an estimated 83,158 deaths are still preventable. The two main vaccines currently deployed, Rotarix and RotaTeq, both live oral vaccines, have been shown to be less effective in developing countries. In addition, they have been associated with a slight risk of intussusception, and the need for cold chain maintenance limits the accessibility of these vaccines to certain areas, leaving 65% of children worldwide unvaccinated and therefore unprotected. Against this backdrop, here we review the main vaccines under development and the state of the art on potential alternatives.
Collapse
Affiliation(s)
- Roberto Cárcamo-Calvo
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain; (R.C.-C.); (C.M.); (J.B.)
| | - Carlos Muñoz
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain; (R.C.-C.); (C.M.); (J.B.)
| | - Javier Buesa
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain; (R.C.-C.); (C.M.); (J.B.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Jesús Rodríguez-Díaz
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain; (R.C.-C.); (C.M.); (J.B.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Roberto Gozalbo-Rovira
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain; (R.C.-C.); (C.M.); (J.B.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| |
Collapse
|
30
|
Rotavirus in Calves and Its Zoonotic Importance. Vet Med Int 2021; 2021:6639701. [PMID: 33968359 PMCID: PMC8081619 DOI: 10.1155/2021/6639701] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/29/2021] [Accepted: 04/02/2021] [Indexed: 12/24/2022] Open
Abstract
Rotavirus is a major pathogen responsible for diarrheal disease in calves, resulting in loss of productivity and economy of farmers. However, various facets of diarrheal disease caused by rotavirus in calves in the world are inadequately understood, considering that diarrheal disease caused by rotavirus is a vital health problem in calves that interrupts production benefits with reduced weight gain and increased mortality, and its potential for zoonotic spread. The pathological changes made by rotavirus are almost exclusively limited to the small intestine that leads to diarrhea. It is environmentally distributed worldwide and was extensively studied. Reassortment is one of the important mechanisms for generating genetic diversity of rotaviruses and eventually for viral evolution. So, the primary strategy is to reduce the burden of rotavirus infections by practicing early colostrum's feeding in newborn calves, using vaccine, and improving livestock management. Rotaviruses have a wide host range, infecting many animal species as well as humans. As it was found that certain animal rotavirus strains had antigenic similarities to some human strains, this may be an indication for an animal to play a role as a source of rotavirus infection in humans. Groups A to C have been shown to infect both humans and animals. The most commonly detected strains in both human and animals are G2, G3, G4, and G9, P [6]. Therefore, this review was made to get overview epidemiology status and zoonotic importance of bovine rotavirus.
Collapse
|
31
|
Buchy P, Chen J, Zhang XH, Benninghoff B, Lee C, Bibera GL. A review of rotavirus vaccine use in Asia and the Pacific regions: challenges and future prospects. Expert Rev Vaccines 2021; 20:1499-1514. [PMID: 33275065 DOI: 10.1080/14760584.2020.1853532] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Rotavirus infection causes a significant proportion of diarrhea disease burden in children <5 years of age in Asia and the Pacific regions. The World Health Organization recommends that rotavirus vaccination should be included in national immunization programs to prevent rotavirus gastroenteritis (RVGE).Areas covered: A literature review was performed to identify and summarize published evidence on RVGE epidemiology and status of rotavirus vaccine use, including the impact and cost-effectiveness of rotavirus vaccination programs in Asia and the Pacific regions (49 countries) during the period 2000-2018.Expert opinion: Rotavirus vaccination programs have successfully reduced the burden of RVGE in many countries. However, such programs still do not exist in most Asia-Pacific countries, and therefore the burden of RVGE remains high in children <5 years of age. Challenges to vaccine implementation include a lack of surveillance data; safety concerns around intussusception; a general lack of awareness about RVGE disease epidemiology and vaccines among physicians, policy-makers, and parents; insufficient cost-effectiveness analyses; and potential issues with vaccine affordability including vaccination costs and lack of political will. Recommendations to overcome these challenges include developing cost-effectiveness analyses for more diverse national and regional settings, providing non-governmental support for low-income countries, and improving advocacy efforts.Plain language summaryWhat is the context?• Rotavirus (RV) infection causes acute gastroenteritis (GE) in children under 5 years of age.• Rotavirus vaccination (RVV) implementation has been slow in Asia and the Pacific (AP) regions, which could be responsible for the region falling behind in their fight against RVGE.What is new?• RVV via national immunization programs (NIPs) is available in 8/49 countries and through the private market or non-governmental support in other countries. Coverage rates vary between countries, possibly driven by the mechanism through which RVV is available.• A substantial positive impact of RVV on RVGE disease burden with a very low risk of intestinal intussusception for up to 7 days after RVV has been documented in the AP regions.• Economic evaluation studies, mainly cost-effectiveness analyses, predict a significant reduction in treatment costs related to RVGE and its complications showing that RVV is good value for money.What is the impact?• The prospect of continued safe and effective use of RVV in the AP regions is promising.• Challenges to RVV implementation include establishing evidence of burden of disease, poor awareness of rotavirus vaccines, limited evidence from cost-effectiveness analyses from several countries, issues of affordability of the vaccine and a lack of political will.• Recommendations for RVV implementation into the NIPs include conducting clinical and cost-effectiveness studies in countries where these are not available, establishing reliable surveillance mechanisms, providing non-governmental support for low-income countries and improving advocacy efforts.• Maintenance of high vaccination coverage is needed in countries that have implemented national RVV programs.Graphical abstract[Formula: see text].
Collapse
|
32
|
Latyshev OE, Eliseeva OV, Kostina LV, Alekseev KP, Khametova KM, Altaeva EG, Verkhovsky OA, Aliper TI, Grebennikova TV. [Assessment of immunogenic activity of the cloned human rotavirus A WA strain.]. Vopr Virusol 2021; 64:156-164. [PMID: 32163681 DOI: 10.36233/0507-4088-2019-64-4-156-164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/10/2019] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Rotovirus infection (RVI) caused by the dsRNA-containing virus from genus Rotavirus, Reoviridae family, belonging to group A (RVA), is the cause of severe diarrhea in human and other mammalian species. Vaccination is the most effective way to reduce the incidence of RVI. At present, the effectiveness of using gnotobiotic piglets as a universal model for reproducing human rotavirus infection and assessing the quality of RVI vaccine preparations has been experimentally proven. OBJECTIVES Evaluation of immunogenic activity of the cloned RVA Wa strain in the new-born Vietnamese potbellied piglets trial. MATERIAL AND METHODS Development of viral preparations of the cloned human Wa strain PBA, development of human RVA rVP6, ELISA, polymerase chain reaction with reverse transcription, immunization and experimental infection of newborn piglets. RESULTS The article presents the results of the experiment on double immunization of newborn piglets with native virus preparations with the infection activity 5.5 lg TCID50/ml, 3 cm3 per dose, HRV with adjuvant 500 µg per dose and mock preparation (control group) followed with experimental inoculation of all animals with virulent virus strain Wa G1P[8] human RVA with infectious activity of 5.5 lg TCID50/ml in 5 cm3 dose. Development of clinical signs of disease and animal death were observed only in control group. RT-PCR system to detect RVA RNA in rectal swabs, samples of small intestine and peripheral lymph nodes was developed. ELISA based on obtained human RVA rVP6 was developed and results on RVA-specific IgG-antibodies in serum samples of experimental piglets are presented. CONCLUSION In the course of the research, a high immunogenic activity of the native and purified virus of the cloned Wa RVA strain Wa was established and the possibility of its use as the main component of the RVI vaccine was confirmed. The possibility of using conventional newborn pigs instead of gnotobiotic piglets as an experimental model was demonstrated.
Collapse
Affiliation(s)
- O E Latyshev
- National Research Center for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russian Federation
| | - O V Eliseeva
- National Research Center for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russian Federation
| | - L V Kostina
- National Research Center for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russian Federation
| | - K P Alekseev
- National Research Center for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russian Federation
| | - K M Khametova
- National Research Center for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russian Federation
| | - E G Altaeva
- Diagnostics and Prevention Research Institute for Human and Animal Diseases, Moscow, 123098, Russian Federation
| | - O A Verkhovsky
- Diagnostics and Prevention Research Institute for Human and Animal Diseases, Moscow, 123098, Russian Federation
| | - T I Aliper
- National Research Center for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russian Federation
| | - T V Grebennikova
- National Research Center for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russian Federation.,Peoples Frendship University of Russia, Moscow, 117198, Russian Federation
| |
Collapse
|
33
|
Shane AL, Weinberg GA. Can We Further Increase Protection Against Rotavirus by Reducing 2 Barriers to Immunization, Inpatient Hospitalization and Older Age? J Pediatric Infect Dis Soc 2021; 10:68-70. [PMID: 31318433 DOI: 10.1093/jpids/piz046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 06/06/2019] [Indexed: 11/13/2022]
Affiliation(s)
- Andi L Shane
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Georgia
| | - Geoffrey A Weinberg
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, New York
| |
Collapse
|
34
|
Groome MJ, Tate JE, Arnold M, Chitnis M, Cox S, de Vos C, Kirsten M, le Grange SM, Loveland J, Machaea S, Maharaj A, Andrews N, Madhi SA, Parashar UD. Evaluation of Intussusception After Oral Monovalent Rotavirus Vaccination in South Africa. Clin Infect Dis 2021; 70:1606-1612. [PMID: 31125061 PMCID: PMC7146001 DOI: 10.1093/cid/ciz431] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/22/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Postlicensure studies have shown an association between rotavirus vaccination and intussusception. We assessed the risk of intussusception associated with Rotarix (RV1) administration, at 6 and 14 weeks of age, in an upper-middle-income country, South Africa. METHODS Active prospective surveillance for intussusception was conducted in 8 hospitals from September 2013 through December 2017. Retrospective case enrollment was done at 1 hospital from July 2012 through August 2013. Demographic characteristics, symptom onset, and rotavirus vaccine status were ascertained. Using the self-controlled case-series method, we estimated age-adjusted incidence rate ratios within 1-7, 8-21, and 1-21 days of rotavirus vaccination in children aged 28-275 days at onset of symptoms. In addition, age-matched controls were enrolled for a subset of cases (n = 169), and a secondary analysis was performed. RESULTS Three hundred forty-six cases were included in the case-series analysis. Post-dose 1, there were zero intussusception cases within 1-7 days, and 5 cases within 8-21 days of vaccination. Post-dose 2, 15 cases occurred within 1-7 days, and 18 cases within 8-21 days of vaccination. There was no increased risk of intussusception 1-7 days after dose 1 (no cases observed) or dose 2 (relative incidence [RI], 1.71 [95% confidence interval {CI} .83-3.01]). Similarly, there was no increased risk 8-21 days after the first (RI, 4.01 [95% CI, .87-10.56]) or second dose (RI, .96 [95% CI, .52-1.60]). Results were similar for the case-control analysis. CONCLUSIONS The risk of intussusception in the 21 days after the first or second dose of RV1 was not higher than the background risk among South Africa infants. CLINICAL TRIALS REGISTRATION South African National Clinical Trial Register (DOH-27-0913-4183).
Collapse
Affiliation(s)
- Michelle J Groome
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences.,Department of Science and Technology/National Research Foundation, Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Marion Arnold
- Red Cross War Memorial Children's Hospital, University of Cape Town.,Tygerberg Hospital, University of Stellenbosch, Cape Town
| | | | - Sharon Cox
- Red Cross War Memorial Children's Hospital, University of Cape Town
| | - Corné de Vos
- Tygerberg Hospital, University of Stellenbosch, Cape Town
| | - Mari Kirsten
- Steve Biko Academic Hospital/Kalafong Hospital, University of Pretoria
| | | | - Jerome Loveland
- Chris Hani Baragwanath Academic Hospital/Charlotte Maxeke Johannesburg Academic Hospital, University of the Witwatersrand, Johannesburg
| | - Sello Machaea
- East London Hospital Complex, Walter Sisulu University
| | - Ashwini Maharaj
- Inkosi Albert Luthuli Hospital, University of Kwa-Zulu Natal, Durban, South Africa
| | - Nick Andrews
- Statistics, Modelling and Economics Department, Public Health England, London, United Kingdom
| | - Shabir A Madhi
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences.,Department of Science and Technology/National Research Foundation, Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | | |
Collapse
|
35
|
Risk of intussusception after monovalent rotavirus vaccine (Rotavac) in Indian infants: A self-controlled case series analysis. Vaccine 2020; 39:78-84. [PMID: 32972735 PMCID: PMC7738754 DOI: 10.1016/j.vaccine.2020.09.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 11/24/2022]
Abstract
Background An association between rotavirus vaccination and intussusception has been documented in post-licensure studies in some countries. We evaluated the risk of intussusception associated with monovalent rotavirus vaccine (Rotavac) administered at 6, 10 and 14 weeks of age in India. Methods Active prospective surveillance for intussusception was conducted at 22 hospitals across 16 states from April 2016 through September 2017. Data on demography, clinical features and vaccination were documented. Age-adjusted relative incidence for 1–7, 8–21, and 1–21 days after rotavirus vaccination in children aged 28–364 days at intussusception onset was estimated using the self-controlled case-series (SCCS) method. Only Brighton Collaboration level 1 cases were included. Results Out of 670 children aged 2–23 months with intussusception, 311 (46.4%) children were aged 28–364 days with confirmed vaccination status. Out of these, 52 intussusception cases with confirmed receipt of RVV were included in the SCCS analysis. No intussusception case was observed within 21 days of dose 1. Only one case occurred during 8–21 days after the dose 2. Post-dose 3, two cases in 1–7 days and 7 cases during 8–21 days period were observed. There was no increased risk of intussusception during 1–7 days after the doses 1 and 2 (zero cases observed) or dose 3 (relative incidence [RI], 1.71 [95% confidence interval {CI} 0.0–5.11]). Similarly, no increased risk during 8–21 days after the dose 1 (zero cases observed), dose 2 (RI, 0.71 [95% CI, 0.0–3.28]) or dose 3 (RI, 2.52 [95% CI, 0.78–5.61]). The results were similar for 1–21 day periods after the doses separately or pooled. Conclusions The risk of intussusception during the first 21 days after any dose of rotavirus vaccine (Rotavac) was not higher among the Indian infants than the background risk, based on limited SCCS analysis of 52 children.
Collapse
|
36
|
Huang WT, Juan YC, Liu CH, Yang YY, Chan KA. Intussusception and Kawasaki disease after rotavirus vaccination in Taiwanese infants. Vaccine 2020; 38:6299-6303. [PMID: 32736940 DOI: 10.1016/j.vaccine.2020.07.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Since 2006, two rotavirus vaccines have been licensed in Taiwan, either as a 2- (RV1) or 3-dose (RV5) schedule administered at ages 2, 4, and 6 months. This study assessed the risk of intussusception and Kawasaki disease (KD) associated with rotavirus vaccines among infants. METHODS Cases of intussusception and KD in infants aged less than 365 days were identified from the National Health Insurance databases, from 1 January 2007 through 31 December 2014, using the first-ever ICD-9-CM diagnosis codes. Histories of rotavirus vaccination were obtained from the National Immunization Information System. The modified self-controlled case series design included vaccinated cases, and compared incidence rate ratios (IRRs) between the risk period (postvaccination days 1-21 [intussusception] or days 1-28 [KD]) and control period (ages 0-364 days outside the -14 to +21 [intussusception] or +28 [KD] days of vaccination) by each type and dose of vaccine. Conditional Poisson regression models were adjusted for age using age-in-week (7-day) categorization. RESULTS Overall 2064 intussusception cases and 2079 KD cases were diagnosed in 567,726 recipients (5313 [0.9%] received both RV5 and RV1). An increase in intussusception risk was observed in the 1-7 days (IRR 12.59, 95% confidence interval [CI] 8.07-19.66) and 8-21 days (IRR 1.78, 95% CI 1.00-3.16) post dose 1 of RV1, but not RV5. Risk of KD was higher during the third week post dose 2 of RV5 (IRR 2.33, 95% CI 1.35-4.00), and fourth week post dose 1 of RV1 (IRR 1.98, 95% CI 1.16-3.40). CONCLUSION Our finding of an increased risk of intussusception associated with RV1 in the first week after dose 1 is consistent with results of previous postlicensure studies. Further research should verify a potentially delayed risk of KD after rotavirus vaccination.
Collapse
Affiliation(s)
| | - Yi-Chen Juan
- Health Data Research Center, National Taiwan University, Taipei, Taiwan
| | - Chia-Hung Liu
- Graduate Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Family Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yen-Yun Yang
- Health Data Research Center, National Taiwan University, Taipei, Taiwan
| | - K Arnold Chan
- Health Data Research Center, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
37
|
Hajela N, Chattopadhyay S, Nair GB, Ganguly NK. Intestinal microbiota and vaccine efficacy in children from resource poor settings - potential impact for the usefulness of probiotics? Benef Microbes 2020; 11:319-328. [PMID: 32720834 DOI: 10.3920/bm2019.0155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Developing countries continue to contribute significantly to the global burden of childhood mortality due to infectious diseases. Infections leading to diseases like diarrhoea, pneumonia and meningitis account for millions of deaths annually. Most of these diseases are preventable by vaccination and therefore global vaccination rates have risen substantially with clear benefits. But paradoxically, the vaccines have demonstrated lower immunogenicity in developing countries as compared to their industrialised counterparts. Malnutrition in resource poor settings along with repeated polymicrobial infections at early age are some of the reasons for the differences in vaccine efficacy in different settings. Recent studies indicate that the gastrointestinal microbiota possibly influences maturation of immune system as well as vaccine efficacy. In this review we discuss evidences from in vitro, animal and human studies showing that probiotics can positively modulate gut microbiota composition and exert immunomodulatory effects on the host. We also discuss how they should be evaluated for their ability to improve vaccine performance especially in low resource settings.
Collapse
Affiliation(s)
- N Hajela
- Gut Microbiota and Probiotic Science Foundation (India), M-4, Level one, South Extension Part - Ii, New Delhi 110049, India
| | - S Chattopadhyay
- Rajiv Gandhi Centre for Biotechnology, Thycaud Post, Poojappura, Thiruvananthapuram, Trivandrum, 695014 Kerala, India
| | - G B Nair
- Rajiv Gandhi Centre for Biotechnology, Thycaud Post, Poojappura, Thiruvananthapuram, Trivandrum, 695014 Kerala, India
| | - N K Ganguly
- Institute of Liver and Biliary Science, New Delhi, India
| |
Collapse
|
38
|
Pitzer VE, Bennett A, Bar-Zeev N, Jere KC, Lopman BA, Lewnard JA, Parashar UD, Cunliffe NA. Evaluating strategies to improve rotavirus vaccine impact during the second year of life in Malawi. Sci Transl Med 2020; 11:11/505/eaav6419. [PMID: 31413144 DOI: 10.1126/scitranslmed.aav6419] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 02/08/2019] [Accepted: 07/25/2019] [Indexed: 01/08/2023]
Abstract
Rotavirus vaccination has substantially reduced the incidence of rotavirus-associated gastroenteritis (RVGE) in high-income countries, but vaccine impact and estimated effectiveness are lower in low-income countries for reasons that are poorly understood. We used mathematical modeling to quantify rotavirus vaccine impact and investigate reduced vaccine effectiveness, particularly during the second year of life, in Malawi, where vaccination was introduced in October 2012 with doses at 6 and 10 weeks. We fitted models to 12 years of prevaccination data and validated the models against postvaccination data to evaluate the magnitude and duration of vaccine protection. The observed rollout of vaccination in Malawi was predicted to lead to a 26 to 77% decrease in the overall incidence of moderate-to-severe RVGE in 2016, depending on assumptions about waning of vaccine-induced immunity and heterogeneity in vaccine response. Vaccine effectiveness estimates were predicted to be higher among 4- to 11-month-olds than 12- to 23-month-olds, even when vaccine-induced immunity did not wane, due to differences in the rate at which vaccinated and unvaccinated individuals acquire immunity from natural infection. We found that vaccine effectiveness during the first and second years of life could potentially be improved by increasing the proportion of infants who respond to vaccination or by lowering the rotavirus transmission rate. An additional dose of rotavirus vaccine at 9 months of age was predicted to lead to higher estimated vaccine effectiveness but to only modest (5 to 16%) reductions in RVGE incidence over the first 3 years after introduction, regardless of assumptions about waning of vaccine-induced immunity.
Collapse
Affiliation(s)
- Virginia E Pitzer
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA.
| | - Aisleen Bennett
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre 3, Malawi.,Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool L69 3BX, UK
| | - Naor Bar-Zeev
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre 3, Malawi.,International Vaccine Access Center, Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Khuzwayo C Jere
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre 3, Malawi.,Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool L69 3BX, UK.,Department of Medical Laboratory Sciences, College of Medicine, University of Malawi, Blantyre 3, Malawi
| | - Benjamin A Lopman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA.,Epidemiology Branch, Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329-4027, USA
| | - Joseph A Lewnard
- Division of Epidemiology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Umesh D Parashar
- Epidemiology Branch, Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329-4027, USA
| | - Nigel A Cunliffe
- Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool L69 3BX, UK
| |
Collapse
|
39
|
Godfrey O, Zhang W, Amponsem-Boateng C, Bonney Oppong T, Zhao Q, Li D. Evidence of rotavirus vaccine impact in sub-Saharan Africa: Systematic review and meta-analysis. PLoS One 2020; 15:e0232113. [PMID: 32339187 PMCID: PMC7185587 DOI: 10.1371/journal.pone.0232113] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/07/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Over 34 countries in Africa have introduced rotavirus vaccine to their national immunization programs: monovalent (Rotarix®, RV1) and pentavalent (RotaTeq®, RV5) after South Africa introduced it in 2009. Since then several studies assessing the impact of the vaccine have been conducted. The principal aim of this study was to evaluate the impact of rotavirus vaccine in sub-Saharan Africa. METHODS A Literature search was performed using Mendeley, PubMed, ScienceDirect, grey literature and Web of Science databases of published studies from January 1, 2017, as years of recent publications on rotavirus vaccine impact in sub-Saharan Africa. A meta-analysis was conducted for rotavirus infection in children under 5 years using proportions of pre and post-vaccine introduction in these populations. Random-effect estimates were considered since the samples were from universal populations. RESULTS Out of the 935 articles identified, 17 studies met the inclusion for systematic review and meta-analysis. The pooled proportion for pre-vaccination period was 42%, 95% (CI: 38-46%), and reduced to 21%, 95% (CI: 17-25%) during post-vaccination period. Rotavirus diarrhea significantly reduced in children < 12 months as compared to children 12-24 months old. Seasonal peaks of rotavirus diarrhea were between June-September. However, data is limited to one year of post-vaccine introduction, and bias may present due to early vaccine impact. CONCLUSION We observed that the introduction of the rotavirus vaccine was partly responsible for the significant reduction in the burden of rotavirus-associated diarrhea in sub-Saharan Africa. Therefore, there is a need to encourage the remaining countries to introduce the vaccine to their routine national immunization programs.
Collapse
Affiliation(s)
- Opolot Godfrey
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Weidong Zhang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Cecilia Amponsem-Boateng
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Timothy Bonney Oppong
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - QingLin Zhao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Dankang Li
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| |
Collapse
|
40
|
Sartorio MUA, Folgori L, Zuccotti G, Mameli C. Rotavirus vaccines in clinical development: Current pipeline and state-of-the-art. Pediatr Allergy Immunol 2020; 31 Suppl 24:58-60. [PMID: 32017224 DOI: 10.1111/pai.13167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 11/28/2022]
Abstract
Rotavirus (RV) disease is a leading cause of mortality and morbidity, especially in children under 5 years of age. The introduction of the two oral rotavirus vaccines Rotarix® and RotaTeq® has shown significant reductions in RV-related mortality, severe RV disease, and hospitalizations. However, some barriers, including a reduced efficacy in low-income countries, safety issues regarding the intussusception risk, age restrictions on vaccine use, the live-attenuated nature itself, and the substantial vaccine costs, currently restrict the full potential of RV disease prevention. Therefore, research is now focusing on the implementation of new oral vaccines and the development of parenteral vaccines to overcome these limits. This review provides an overview of the new rotavirus vaccines in clinical development and the ongoing clinical trials on new RV vaccines in the pediatric age.
Collapse
Affiliation(s)
| | - Laura Folgori
- Paediatric Infectious Disease Unit, Department of Pediatrics, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, V. Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Chiara Mameli
- Department of Pediatrics, V. Buzzi Children's Hospital, University of Milan, Milan, Italy
| |
Collapse
|
41
|
de Ruiter K, Jochems SP, Tahapary DL, Stam KA, König M, van Unen V, Laban S, Höllt T, Mbow M, Lelieveldt BPF, Koning F, Sartono E, Smit JWA, Supali T, Yazdanbakhsh M. Helminth infections drive heterogeneity in human type 2 and regulatory cells. Sci Transl Med 2020; 12:12/524/eaaw3703. [DOI: 10.1126/scitranslmed.aaw3703] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 08/06/2019] [Accepted: 12/03/2019] [Indexed: 01/03/2023]
Abstract
Helminth infections induce strong type 2 and regulatory responses, but the degree of heterogeneity of such cells is not well characterized. Using mass cytometry, we profiled these cells in Europeans and Indonesians not exposed to helminths and in Indonesians residing in rural areas infected with soil-transmitted helminths. To assign immune alteration to helminth infection, the profiling was performed before and 1 year after deworming. Very distinct signatures were found in Europeans and Indonesians, showing expanded frequencies of T helper 2 cells, particularly CD161+ cells and ILC2s in helminth-infected Indonesians, which was confirmed functionally through analysis of cytokine-producing cells. Besides ILC2s and CD4+ T cells, CD8+ T cells and γδ T cells in Indonesians produced type 2 cytokines. Regulatory T cells were also expanded in Indonesians, but only those expressing CTLA-4, and some coexpressed CD38, HLA-DR, ICOS, or CD161. CD11c+ B cells were found to be the main IL-10 producers among B cells in Indonesians, a subset that was almost absent in Europeans. A number of the distinct immune profiles were driven by helminths as the profiles reverted after clearance of helminth infections. Moreover, Indonesians with no helminth infections residing in an urban area showed immune profiles that resembled Europeans rather than rural Indonesians, which excludes a major role for ethnicity. Detailed insight into the human type 2 and regulatory networks could provide opportunities to target these cells for more precise interventions.
Collapse
Affiliation(s)
- Karin de Ruiter
- Department of Parasitology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Simon P. Jochems
- Department of Parasitology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Dicky L. Tahapary
- Department of Parasitology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
- Department of Internal Medicine, Division of Endocrinology, Dr. Cipto Mangunkusumo National General Hospital, Faculty of Medicine Universitas Indonesia, 10430 Jakarta, Indonesia
- Metabolic, Cardiovascular and Aging Cluster, The Indonesian Medical Education and Research Institute, Universitas Indonesia, 10430 Jakarta, Indonesia
| | - Koen A. Stam
- Department of Parasitology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Marion König
- Department of Parasitology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Vincent van Unen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Sandra Laban
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Thomas Höllt
- Computer Graphics and Visualization Group, Delft University of Technology, 2628 XE Delft, Netherlands
- Computational Biology Center, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Moustapha Mbow
- Department of Immunology, Cheikh Anta Diop University of Dakar (UCAD), 5005 Dakar, Senegal
| | - Boudewijn P. F. Lelieveldt
- Department of LKEB Radiology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
- Department of Pattern Recognition and Bioinformatics Group, Delft University of Technology, 2628 XE Delft, Netherlands
| | - Frits Koning
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Erliyani Sartono
- Department of Parasitology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Johannes W. A. Smit
- Department of Internal Medicine, Radboud University Medical Centre, 6525 GA Nijmegen, Netherlands
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Taniawati Supali
- Department of Parasitology, Faculty of Medicine Universitas Indonesia, 10430 Jakarta, Indonesia
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| |
Collapse
|
42
|
Motamedi-Rad M, Farahmand M, Arashkia A, Jalilvand S, Shoja Z. VP7 and VP4 genotypes of rotaviruses cocirculating in Iran, 2015 to 2017: Comparison with cogent sequences of Rotarix and RotaTeq vaccine strains before their use for universal mass vaccination. J Med Virol 2019; 92:1110-1123. [PMID: 31774174 DOI: 10.1002/jmv.25642] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 11/23/2019] [Indexed: 12/17/2022]
Abstract
The present study was conducted to analyze the genotypic diversity of circulating species A rotavirus (RVA) strains in Iran and also to investigate comparative analysis between the genotypes of VP4 and VP7 of cocirculating RVA and vaccine strains before the vaccine is introduced in the national immunization program. The G3-lineage I was found in this study as the most common G genotype which was followed by G9-lineage III, G1-lineages I, II, G12-lineage III, G2-lineage IV, and G4-lineage I. Also, P[8]-lineages III, IV was found as the predominant P genotype which was followed by P[4]-lineage V, and P[6]-lineage I. Overally, G3P[8] was determined as the most common combination. Moreover, the analysis of the VP7 antigenic epitopes showed that several amino acid differences existed between circulating Iranian and the vaccine strains. The comparison of genotype G1 of Iranian and vaccine strains (RotaTeq and Rotarix), and genotypes G2, G3, and G4 of Iranian and RotaTeq vaccine strains revealed three to five amino acids differences on the VP7 antigenic epitopes. Furthermore, analyzing of the VP8* epitopes of Iranian P[8] strains indicated that they contained up to 11 and 14 amino acid differences with Rotarix and RotaTeq, respectively. Based on different patterns of amino acid substitutions in circulating and vaccine strains, the emergence of antibody escaping mutants and potentially the decrease of immune protection might ensue in vaccinated children. However, considering the broad cross-protective activity of RVA vaccines, their efficacy should be monitored after the introduction in Iran.
Collapse
Affiliation(s)
| | - Mohammad Farahmand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Arashkia
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Somayeh Jalilvand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Zabihollah Shoja
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
43
|
Enteropathogens and Rotavirus Vaccine Immunogenicity in a Cluster Randomized Trial of Improved Water, Sanitation and Hygiene in Rural Zimbabwe. Pediatr Infect Dis J 2019; 38:1242-1248. [PMID: 31738342 PMCID: PMC7205402 DOI: 10.1097/inf.0000000000002485] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Oral rotavirus vaccines (RVVs) are less efficacious in low-income versus high-income settings, plausibly due to more enteropathogen exposure through poor water, sanitation and hygiene (WASH). We explored associations between enteropathogens and RVV immunogenicity and evaluated the effect of improved WASH on enteropathogen carriage. METHODS We detected stool enteropathogens using quantitative molecular methods and measured anti-rotavirus immunoglobulin A by enzyme-linked immunosorbent assay in infants enrolled to a cluster randomized 2 × 2 factorial trial of improved WASH and improved infant feeding in Zimbabwe (NCT01824940). We used multivariable regression to explore associations between enteropathogens and RVV seroconversion, seropositivity and geometric mean titer. We evaluated effects of improved WASH on enteropathogen prevalence using linear and binomial regression models with generalized estimating equations. RESULTS Among 224 infants with enteropathogen and immunogenicity data, 107 (47.8%) had ≥1 pathogen and 39 (17.4%) had ≥2 pathogens detected at median age 41 days (interquartile range: 35-54). RVV seroconversion was low (23.7%). After adjusting for Sabin-poliovirus quantity, pan-enterovirus quantity was positively associated with RVV seroconversion (relative risk 1.61 per 10-fold increase in pan-enterovirus; 95% confidence interval: 1.35-1.91); in the same model, Sabin quantity was negatively associated with RVV seroconversion (relative risk: 0.76; 95% confidence interval: 0.60-0.96). There were otherwise no meaningful associations between individual or total pathogens (bacteria, viruses, parasites or all pathogens) and any measure of RVV immunogenicity. Enteropathogen detection did not differ between randomized WASH and non-WASH groups. CONCLUSIONS Enteropathogen infections were common around the time of rotavirus vaccination in rural Zimbabwean infants but did not explain poor RVV immunogenicity and were not reduced by a package of household-level WASH interventions.
Collapse
|
44
|
Antirotavirus IgA seroconversion rates in children who receive concomitant oral poliovirus vaccine: A secondary, pooled analysis of Phase II and III trial data from 33 countries. PLoS Med 2019; 16:e1003005. [PMID: 31887139 PMCID: PMC6936798 DOI: 10.1371/journal.pmed.1003005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/22/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Despite the success of rotavirus vaccines over the last decade, rotavirus remains a leading cause of severe diarrheal disease among young children. Further progress in reducing the burden of disease is inhibited, in part, by vaccine underperformance in certain settings. Early trials suggested that oral poliovirus vaccine (OPV), when administered concomitantly with rotavirus vaccine, reduces rotavirus seroconversion rates after the first rotavirus dose with modest or nonsignificant interference after completion of the full rotavirus vaccine course. Our study aimed to identify a range of individual-level characteristics, including concomitant receipt of OPV, that affect rotavirus vaccine immunogenicity in high- and low-child-mortality settings, controlling for individual- and country-level factors. Our central hypothesis was that OPV administered concomitantly with rotavirus vaccine reduced rotavirus vaccine immunogenicity. METHODS AND FINDINGS Pooled, individual-level data from GlaxoSmithKline's Phase II and III clinical trials of the monovalent rotavirus vaccine (RV1), Rotarix, were analyzed, including 7,280 vaccinated infants (5-17 weeks of age at first vaccine dose) from 22 trials and 33 countries/territories (5 countries/territories with high, 13 with moderately low, and 15 with very low child mortality). Two standard markers for immune response were examined including antirotavirus immunoglobulin A (IgA) seroconversion (defined as the appearance of serum antirotavirus IgA antibodies in subjects initially seronegative) and serum antirotavirus IgA titer, both collected approximately 4-12 weeks after administration of the last rotavirus vaccine dose. Mixed-effect logistic regression and mixed-effect linear regression of log-transformed data were used to identify individual- and country-level predictors of seroconversion (dichotomous) and antibody titer (continuous), respectively. Infants in high-child-mortality settings had lower odds of seroconverting compared with infants in low-child-mortality settings (odds ratio [OR] = 0.48, 95% confidence interval [CI] 0.43-0.53, p < 0.001). Similarly, among those who seroconverted, infants in high-child-mortality settings had lower IgA titers compared with infants in low-child-mortality settings (mean difference [β] = 0.83, 95% CI 0.77-0.90, p < 0.001). Infants who received OPV concomitantly with both their first and their second doses of rotavirus vaccine had 0.63 times the odds of seroconverting (OR = 0.63, 95% CI 0.47-0.84, p = 0.002) compared with infants who received OPV but not concomitantly with either dose. In contrast, among infants who seroconverted, OPV concomitantly administered with both the first and second rotavirus vaccine doses was found to be positively associated with antirotavirus IgA titer (β = 1.28, 95% CI 1.07-1.53, p = 0.009). Our findings may have some limitations in terms of generalizability to routine use of rotavirus vaccine because the analysis was limited to healthy infants receiving RV1 in clinical trial settings. CONCLUSIONS Our findings suggest that OPV given concomitantly with RV1 was a substantial contributor to reduced antirotavirus IgA seroconversion, and this interference was apparent after the second vaccine dose of RV1, as with the original clinical trials that our reanalysis is based on. However, our findings do suggest that the forthcoming withdrawal of OPV from the infant immunization schedule globally has the potential to improve RV1 performance.
Collapse
|
45
|
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
|
46
|
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
|
47
|
Lewnard JA, Lopman BA, Parashar UD, Bennett A, Bar-Zeev N, Cunliffe NA, Samuel P, Guerrero ML, Ruiz-Palacios G, Kang G, Pitzer VE. Heterogeneous susceptibility to rotavirus infection and gastroenteritis in two birth cohort studies: Parameter estimation and epidemiological implications. PLoS Comput Biol 2019; 15:e1007014. [PMID: 31348775 PMCID: PMC6690553 DOI: 10.1371/journal.pcbi.1007014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 08/12/2019] [Accepted: 04/09/2019] [Indexed: 11/19/2022] Open
Abstract
Cohort studies, randomized trials, and post-licensure studies have reported reduced natural and vaccine-derived protection against rotavirus gastroenteritis (RVGE) in low- and middle-income countries. While susceptibility of children to rotavirus is known to vary within and between settings, implications for estimation of immune protection are not well understood. We sought to re-estimate naturally-acquired protection against rotavirus infection and RVGE, and to understand how differences in susceptibility among children impacted estimates. We re-analyzed data from studies conducted in Mexico City, Mexico and Vellore, India. Cumulatively, 573 rotavirus-unvaccinated children experienced 1418 rotavirus infections and 371 episodes of RVGE over 17,636 child-months. We developed a model that characterized susceptibility to rotavirus infection and RVGE among children, accounting for aspects of the natural history of rotavirus and differences in transmission rates between settings. We tested whether model-generated susceptibility measurements were associated with demographic and anthropometric factors, and with the severity of RVGE symptoms. We identified greater variation in susceptibility to rotavirus infection and RVGE in Vellore than in Mexico City. In both cohorts, susceptibility to rotavirus infection and RVGE were associated with male sex, lower birth weight, lower maternal education, and having fewer siblings; within Vellore, susceptibility was also associated with lower socioeconomic status. Children who were more susceptible to rotavirus also experienced higher rates of rotavirus-negative diarrhea, and higher risk of moderate-to-severe symptoms when experiencing RVGE. Simulations suggested that discrepant estimates of naturally-acquired immunity against RVGE can be attributed, in part, to between-setting differences in susceptibility of children, but result primarily from the interaction of transmission rates with age-dependent risk for infections to cause RVGE. We found that more children in Vellore than in Mexico City belong to a high-risk group for rotavirus infection and RVGE, and demonstrate that unmeasured individual- and age-dependent susceptibility may influence estimates of naturally-acquired immune protection against RVGE.
Collapse
Affiliation(s)
- Joseph A. Lewnard
- Division of Epidemiology and Biostatistics, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Benjamin A. Lopman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Umesh D. Parashar
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Aisleen Bennett
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
- Center for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, University of Liverpool, Liverpool, United Kingdom
| | - Naor Bar-Zeev
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
- Center for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, University of Liverpool, Liverpool, United Kingdom
- International Vaccine Access Center, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Nigel A. Cunliffe
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
- Center for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, University of Liverpool, Liverpool, United Kingdom
| | - Prasanna Samuel
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - M. Lourdes Guerrero
- Instituto Nacional de Ciences Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Gagandeep Kang
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - Virginia E. Pitzer
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| |
Collapse
|
48
|
Ella R, Babji S, Ciarlet M, Blackwelder WC, Vadrevu KM. A randomized, open-labelled, non-inferiority phase 4 clinical trial to evaluate the immunogenicity and safety of the live, attenuated, oral rotavirus vaccine, ROTAVAC® in comparison with a licensed rotavirus vaccine in healthy infants. Vaccine 2019; 37:4407-4413. [PMID: 31178377 DOI: 10.1016/j.vaccine.2019.05.069] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/11/2019] [Accepted: 05/06/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND ROTAVAC® (nHRV), derived naturally from the human 116E rotavirus (RV) neonatal strain, was licensed in India in 2015 based on promising results of a phase 3, safety and efficacy vaccine trial. As a pre-requisite for WHO prequalification, we compared the immunogenicity and safety of ROTAVAC® to those of a WHO-prequalified, Rotarix®. METHODS We conducted a multicentre, open-labeled, randomized phase 4 clinical trial where 464 infants, 6-8 weeks of age were equally randomized to receive as licensed, the complete regimen of ROTAVAC® (3 doses; Group I) or Rotarix® (2 doses; Group II). Antibody responses (serum anti-RV Immunoglobulin A [IgA]) were measured by enzyme-linked immunosorbent assay (ELISA). The primary analysis was an assessment of non-inferiority of ROTAVAC® to Rotarix® for geometric mean concentration (GMC) for infants who received the complete regimen of either vaccine. RESULTS The GMC for Group I was 20.4 (95%CI: 17.6, 23.6) and that for Group II was 24.8 (95%CI: 20.3, 30.3), the GMC ratio was 0.82 (95% CI: 0.64, 1.05), thus meeting the non-inferiority criterion. Site-wise analysis of GMC titres revealed that one site had a peculiar pre-vaccination titre affecting only ROTAVAC® post-vaccination GMCs. Seroconversion rates were 35.3% (95%CI: 29.0, 41.9) and 31.0% (95%CI: 25.1, 37.4) for Groups I and Group II, respectively. There was no substantive difference in safety profiles between both vaccines. CONCLUSIONS The complete regimen of ROTAVAC® demonstrated immunological non-inferiority to the complete regimen of Rotarix® with a clinically acceptable safety profile. Because the demand for RV vaccines is increasing as more countries are expanding their immunization schedules, the lack of need of a buffering agent, low dose volume (0.5 mL), non-interference with other concomitantly administered vaccines, and conformance with WHO-prequalification requirements provide ROTAVAC® the potential for widespread global usage. Post completion of this study, ROTAVAC® is now a WHO-prequalified vaccine. CLINICAL TRIALS REGISTRATION (CTRI Number: CTRI/2015/12/006428).
Collapse
Affiliation(s)
- Raches Ella
- Bharat Biotech International Limited, Genome Valley, Shameerpet, Hyderabad, India
| | - Sudhir Babji
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - Max Ciarlet
- Independent Clinical Development Consultant, USA
| | | | | |
Collapse
|
49
|
Polio endgame: Lessons for the global rotavirus vaccination program. Vaccine 2019; 37:3040-3049. [DOI: 10.1016/j.vaccine.2019.04.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 04/02/2019] [Accepted: 04/08/2019] [Indexed: 12/19/2022]
|
50
|
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
|