1
|
Tahrat H, Munir A, Marchetti F. Rotavirus vaccine coverage, completion, and compliance: A systematic literature review. Hum Vaccin Immunother 2025; 21:2442780. [PMID: 39751000 DOI: 10.1080/21645515.2024.2442780] [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: 09/26/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Rotavirus, a leading cause of severe acute gastroenteritis in children, is largely preventable through immunization with two internationally licensed oral rotavirus vaccines (RVVs) included in national programs across over 100 countries. These RVVs are administered in either two (Rotarix™; 2D-RV) or three (RotaTeq®; 3D-RV) doses. We aimed to assess the global coverage, completion, and compliance of 2D-RV and 3D-RV in various settings, and to identify factors influencing vaccine coverage. We conducted a systematic review of PubMed and Embase for articles published between 2006 and 2021. We included 74 publications across 31 countries. RVV coverage rates and the factors associated with coverage varied widely among countries based on income level, RVV used, and the year of vaccination. Due to market bias and insufficient studies, valid RVVs coverage comparisons couldn't be made. However, 2D-RV had better completion/compliance rates than 3D-RV in Italy, Mexico, and the US.
Collapse
|
2
|
Wandera EA, Kurokawa N, Mutua MM, Muriithi B, Nyangao J, Khamadi SA, Kathiiko C, Wachira M, Njuguna E, Mwaura B, Golicha RO, Njau J, Morita K, Kaneko S, Komoto S, Tsutsui N, Ichinose Y. Long-term impact of rotavirus vaccination on all-cause and rotavirus-specific gastroenteritis and strain distribution in Central Kenya: An 11-year interrupted time-series analysis. Vaccine 2024; 42:126210. [PMID: 39151233 DOI: 10.1016/j.vaccine.2024.126210] [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: 03/27/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Kenya introduced a monovalent rotavirus vaccine administered orally at 6 and 10 weeks of age into her National Immunization Program in July 2014. The study evaluated the long-term impact of the vaccine on hospitalization for all-cause and rotavirus-specific acute gastroenteritis (AGE) and strain epidemiology in Kenya. METHODS Data on all-cause and rotavirus-specific AGE and strain distribution were derived from an eleven-year hospital-based surveillance of AGE among children aged <5 years at Kiambu County Teaching and Referral Hospital (KCTRH) in Central Kenya between 2009 and 2020. Fecal samples were screened for group A rotavirus using ELISA and genotyped using multiplex semi-nested RT-PCR. Trends in all-cause and rotavirus-related AGE and strain distribution were compared between the pre-vaccine (July 2009-June 2014), early post-vaccine (July 2014-June 2016) and late post-vaccine (February 2019-October 2020) periods. RESULTS Rotavirus-specific AGE was detected at 27.5% (429/1546, 95% CI: 25.5-30.1%) in the pre-vaccine period; 13.8% (91/658, 95% CI: 11.3-16.6%) in the early post-vaccine period (July 2014-June 2016); and 12.0% (229/1916, 95% CI: 10.6-13.5%) in the late post-vaccine period (February 2019-October 2020). This amounted to a decline of 49.8% (95% CI: 34.6%-63.7%) in rotavirus-specific AGE in the early post-vaccine period and 53.4% (95% CI: 41.5-70.3%) in the late post-vaccine period when compared to the pre-vaccine period. All-cause AGE hospitalizations declined by 40.2% (95% CI: 30.8%-50.2%) and 75.3% (95% CI: 65.9-83.1%) in the early post-vaccine and late post-vaccine periods, respectively, when compared to the pre-vaccine period. G3P [8] was the predominant strain in the late post-vaccine period, replacing G1P[8] which had predominated in the pre-vaccine and early post-vaccine periods. Additionally, we detected considerable proportions of uncommon strains G3P[6] (4.8%) and G12P[6] (3.5%) in the post-vaccine era. CONCLUSION Rotavirus vaccination has resulted in a significant decline in all-cause and rotavirus-specific AGE, and thus, provides strong evidence for public health policy makers in Kenya to support the sustained use of the rotavirus vaccine in routine immunization. However, the shift in strain dominance and age distribution of rotavirus AGE in the post-vaccine era underscores the need for continued surveillance to assess any possible vaccine-induced selective pressure that could diminish the vaccine effectiveness over time.
Collapse
Affiliation(s)
- Ernest Apondi Wandera
- Centre for Virus Research, Kenya Medical Research Institute, P.O. Box 54840-00200, Nairobi, Kenya,; Institute of Tropical Medicine, Nagasaki University, P.O. Box 19993-00202, Nairobi, Kenya,.
| | - Natsuki Kurokawa
- Department of Project Planning and Management, Mitsubishi Tanabe Pharma Corporation, 1-1-1, Marunouchi Chiyoda-ku, Tokyo 100-8205, Japan
| | - Maurine Mumo Mutua
- Institute of Tropical Medicine, Nagasaki University, P.O. Box 19993-00202, Nairobi, Kenya
| | - Betty Muriithi
- Institute of Tropical Medicine, Nagasaki University, P.O. Box 19993-00202, Nairobi, Kenya
| | - James Nyangao
- Centre for Virus Research, Kenya Medical Research Institute, P.O. Box 54840-00200, Nairobi, Kenya
| | - Samoel Ashimosi Khamadi
- Centre for Virus Research, Kenya Medical Research Institute, P.O. Box 54840-00200, Nairobi, Kenya
| | - Cyrus Kathiiko
- Institute of Tropical Medicine, Nagasaki University, P.O. Box 19993-00202, Nairobi, Kenya
| | - Mary Wachira
- Institute of Tropical Medicine, Nagasaki University, P.O. Box 19993-00202, Nairobi, Kenya
| | - Eunice Njuguna
- Institute of Tropical Medicine, Nagasaki University, P.O. Box 19993-00202, Nairobi, Kenya
| | - Boniface Mwaura
- Institute of Tropical Medicine, Nagasaki University, P.O. Box 19993-00202, Nairobi, Kenya
| | - Rahma Ordofa Golicha
- Centre for Microbiology Research, Kenya Medical Research Institute, P.O. Box 54840-00200 Nairobi, Kenya
| | - Joseph Njau
- Department of Pediatrics, Kiambu County Teaching and Referral Hospital, P.O. Box 39-00900 Kiambu, Kenya
| | - Kouichi Morita
- Institute of Tropical Medicine, Nagasaki University, P.O. Box 19993-00202, Nairobi, Kenya
| | - Satoshi Kaneko
- Institute of Tropical Medicine, Nagasaki University, P.O. Box 19993-00202, Nairobi, Kenya
| | - Satoshi Komoto
- Department of Virology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho,Toyoake, Aichi 470-1192, Japan,; Division of One Health, Research Center for Global and Local Infectious Diseases, 1-1 Idaigaoka, Hasama-machi, Oita University, Yufu 879-5593, Oita, Japan
| | - Naohisa Tsutsui
- Department of Project Planning and Management, Mitsubishi Tanabe Pharma Corporation, 1-1-1, Marunouchi Chiyoda-ku, Tokyo 100-8205, Japan
| | - Yoshio Ichinose
- Institute of Tropical Medicine, Nagasaki University, P.O. Box 19993-00202, Nairobi, Kenya
| |
Collapse
|
3
|
Ossio A, Flores-Rodríguez F, Heredia N, García S, Merino-Mascorro JA. Foodborne Viruses and Somatic Coliphages Occurrence in Fresh Produce at Retail from Northern Mexico. FOOD AND ENVIRONMENTAL VIROLOGY 2024; 16:109-119. [PMID: 38198031 DOI: 10.1007/s12560-023-09578-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/28/2023] [Indexed: 01/11/2024]
Abstract
Foodborne disease outbreaks linked to consumption of vegetables have been often attributed to human enteric viruses, such as Norovirus (NoV), Hepatitis A virus (HAV), and Rotavirus (RoV). Information about the occurrence of these viruses is scarce in many fresh-producing countries. Viral contamination detection of indicators, such as somatic coliphages, could indirectly reflect the presence of viral pathogens, being a valuable tool for better viral risk assessment in food industry. This study aimed to establish the occurrence and correlation of foodborne viruses and somatic coliphages in leafy greens in northern Mexico. A total of 320 vegetable samples were collected, resulting in 80 composite rinses, 40 of lettuce and 40 of parsley. Somatic coliphages were determined using the EPA 1602 method, while foodborne viruses (HAV, RoV, NoV GI, and GII) were determined by qPCR. The occurrence of RoV was 22.5% (9/40, mean 2.11 log gc/g) in lettuce and 20% (8/40, mean 1.91 log gc/g) in parsley. NoV and HAV were not detected in any samples. Somatic coliphages were present in all lettuce and parsley samples, with mean levels of 1.85 log PFU/100 ml and 2.28 log PFU/100 ml, respectively. Spearman analysis established the correlation of somatic coliphages and genomic copies of RoV, resulting in an r2 value of - 0.026 in lettuce and 0.349 in parsley. Although NoV or HAV were undetected in the samples, the presence of RoV is a matter of concern as leafy greens are usually eaten raw, which poses a potential risk of infection.
Collapse
Affiliation(s)
- Axel Ossio
- Laboratorio de Bioquímica y Genética de Microorganismos, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, 66455, San Nicolas de los Garza, N.L., Mexico
| | - Fernanda Flores-Rodríguez
- Laboratorio de Bioquímica y Genética de Microorganismos, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, 66455, San Nicolas de los Garza, N.L., Mexico
| | - Norma Heredia
- Laboratorio de Bioquímica y Genética de Microorganismos, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, 66455, San Nicolas de los Garza, N.L., Mexico
| | - Santos García
- Laboratorio de Bioquímica y Genética de Microorganismos, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, 66455, San Nicolas de los Garza, N.L., Mexico
| | - Jose Angel Merino-Mascorro
- Laboratorio de Bioquímica y Genética de Microorganismos, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, 66455, San Nicolas de los Garza, N.L., Mexico.
| |
Collapse
|
4
|
Le LKT, Chu MNT, Tate JE, Jiang B, Bowen MD, Esona MD, Gautam R, Jaimes J, Pham TPT, Huong NT, Anh DD, Trang NV, Parashar U. Genetic diversity of G9, G3, G8 and G1 rotavirus group A strains circulating among children with acute gastroenteritis in Vietnam from 2016 to 2021. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 118:105566. [PMID: 38316245 PMCID: PMC11299202 DOI: 10.1016/j.meegid.2024.105566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
Rotavirus group A (RVA) is the most common cause of severe childhood diarrhea worldwide. The introduction of rotavirus vaccination programs has contributed to a reduction in hospitalizations and mortality caused by RVA. From 2016 to 2021, we conducted surveillance to monitor RVA prevalence and genotype distribution in Nam Dinh and Thua Thien Hue (TT Hue) provinces where a pilot Rotavin-M1 vaccine (Vietnam) implementation took place from 2017 to 2020. Out of 6626 stool samples, RVA was detected in 2164 (32.6%) by ELISA. RT-PCR using type-specific primers were used to determine the G and P genotypes of RVA-positive specimens. Whole genome sequences of a subset of 52 specimens randomly selected from 2016 to 2021 were mapped using next-generation sequencing. From 2016 to 2021, the G9, G3 and G8 strains dominated, with detected frequencies of 39%, 23%, and 19%, respectively; of which, the most common genotypes identified were G9P[8], G3P[8] and G8P[8]. G1 strains re-emerged in Nam Dinh and TT Hue (29.5% and 11.9%, respectively) from 2020 to 2021. G3 prevalence decreased from 74% to 20% in TT Hue and from 21% to 13% in Nam Dinh province between 2017 and 2021. The G3 strains consisted of 52% human typical G3 (hG3) and 47% equine-like G3 (eG3). Full genome analysis showed substantial diversity among the circulating G3 strains with different backgrounds relating to equine and feline viruses. G9 prevalence decreased sharply from 2016 to 2021 in both provinces. G8 strains peaked during 2019-2020 in Nam Dinh and TT Hue provinces (68% and 46%, respectively). Most G8 and G9 strains had no genetic differences over the surveillance period with very high nucleotide similarities of 99.2-99.9% and 99.1-99.7%, respectively. The G1 strains were not derived from the RVA vaccine. Changes in the genotype distribution and substantial diversity among circulating strains were detected throughout the surveillance period and differed between the two provinces. Determining vaccine effectiveness against circulating strains over time will be important to ensure that observed changes are due to natural secular variation and not from vaccine pressure.
Collapse
Affiliation(s)
- Ly K T Le
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Viet Nam
| | - Mai N T Chu
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Viet Nam
| | - Jacqueline E Tate
- United States Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Baoming Jiang
- United States Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Michael D Bowen
- United States Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Mathew D Esona
- United States Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Rashi Gautam
- United States Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Jose Jaimes
- United States Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Thao P T Pham
- Center for Research and Production of Vaccines and Biologicals, Hanoi 100000, Viet Nam
| | - Nguyen T Huong
- Center for Research and Production of Vaccines and Biologicals, Hanoi 100000, Viet Nam
| | - Dang D Anh
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Viet Nam
| | - Nguyen V Trang
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Viet Nam.
| | - Umesh Parashar
- United States Centers for Disease Control and Prevention, Atlanta, GA 30333, USA.
| |
Collapse
|
5
|
Colston JM, Fang B, Houpt E, Chernyavskiy P, Swarup S, Gardner LM, Nong MK, Badr HS, Zaitchik BF, Lakshmi V, Kosek MN. The Planetary Child Health & Enterics Observatory (Plan-EO): A protocol for an interdisciplinary research initiative and web-based dashboard for mapping enteric infectious diseases and their risk factors and interventions in LMICs. PLoS One 2024; 19:e0297775. [PMID: 38412156 PMCID: PMC10898779 DOI: 10.1371/journal.pone.0297775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 01/12/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Diarrhea remains a leading cause of childhood illness throughout the world that is increasing due to climate change and is caused by various species of ecologically sensitive pathogens. The emerging Planetary Health movement emphasizes the interdependence of human health with natural systems, and much of its focus has been on infectious diseases and their interactions with environmental and human processes. Meanwhile, the era of big data has engendered a public appetite for interactive web-based dashboards for infectious diseases. However, enteric infectious diseases have been largely overlooked by these developments. METHODS The Planetary Child Health & Enterics Observatory (Plan-EO) is a new initiative that builds on existing partnerships between epidemiologists, climatologists, bioinformaticians, and hydrologists as well as investigators in numerous low- and middle-income countries. Its objective is to provide the research and stakeholder community with an evidence base for the geographical targeting of enteropathogen-specific child health interventions such as novel vaccines. The initiative will produce, curate, and disseminate spatial data products relating to the distribution of enteric pathogens and their environmental and sociodemographic determinants. DISCUSSION As climate change accelerates there is an urgent need for etiology-specific estimates of diarrheal disease burden at high spatiotemporal resolution. Plan-EO aims to address key challenges and knowledge gaps by making and disseminating rigorously obtained, generalizable disease burden estimates. Pre-processed environmental and EO-derived spatial data products will be housed, continually updated, and made publicly available for download to the research and stakeholder communities. These can then be used as inputs to identify and target priority populations living in transmission hotspots and for decision-making, scenario-planning, and disease burden projection. STUDY REGISTRATION PROSPERO protocol #CRD42023384709.
Collapse
Affiliation(s)
- Josh M. Colston
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Bin Fang
- Department of Civil and Environmental Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Eric Houpt
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Pavel Chernyavskiy
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Samarth Swarup
- Biocomplexity Institute, University of Virginia, Charlottesville, Virginia, United States of America
| | - Lauren M. Gardner
- Department of Civil and Systems Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Malena K. Nong
- University of Virginia College of Arts & Sciences, Charlottesville, Virginia, United States of America
| | - Hamada S. Badr
- Department of Earth and Planetary Sciences, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Benjamin F. Zaitchik
- Department of Earth and Planetary Sciences, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Venkataraman Lakshmi
- Department of Civil and Environmental Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Margaret N. Kosek
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| |
Collapse
|
6
|
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
|
7
|
Colston JM, Chernyavskiy P, Gardner L, Nong M, Fang B, Houpt E, Swarup S, Badr H, Zaitchik B, Lakshmi V, Kosek M. The Planetary Child Health & Enterics Observatory (Plan-EO): a protocol for an interdisciplinary research initiative and web-based dashboard for mapping enteric infectious diseases and their risk factors and interventions in LMICs. RESEARCH SQUARE 2024:rs.3.rs-2640564. [PMID: 36993232 PMCID: PMC10055683 DOI: 10.21203/rs.3.rs-2640564/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Background Diarrhea remains a leading cause of childhood illness throughout the world that is increasing due to climate change and is caused by various species of ecologically sensitive pathogens. The emerging Planetary Health movement emphasizes the interdependence of human health with natural systems, and much of its focus has been on infectious diseases and their interactions with environmental and human processes. Meanwhile, the era of big data has engendered a public appetite for interactive web-based dashboards for infectious diseases. However, enteric infectious diseases have been largely overlooked by these developments. Methods The Planetary Child Health and Enterics Observatory (Plan-EO) is a new initiative that builds on existing partnerships between epidemiologists, climatologists, bioinformaticians, and hydrologists as well as investigators in numerous low- and middle-income countries. Its objective is to provide the research and stakeholder community with an evidence base for the geographical targeting of enteropathogen-specific child health interventions such as novel vaccines. The initiative will produce, curate, and disseminate spatial data products relating to the distribution of enteric pathogens and their environmental and sociodemographic determinants. Discussion As climate change accelerates there is an urgent need for etiology-specific estimates of diarrheal disease burden at high spatiotemporal resolution. Plan-EO aims to address key challenges and knowledge gaps by making rigorously obtained, generalizable disease burden estimates freely available and accessible to the research and stakeholder communities. Pre-processed environmental and EO-derived spatial data products will be housed, continually updated, and made publicly available to the research and stakeholder communities both within the webpage itself and for download. These inputs can then be used to identify and target priority populations living in transmission hotspots and for decision-making, scenario-planning, and disease burden projection. Study registration PROSPERO protocol #CRD42023384709.
Collapse
Affiliation(s)
| | | | | | - Malena Nong
- University of Virginia College of Arts & Sciences
| | | | - Eric Houpt
- University of Virginia School of Medicine
| | | | | | | | | | | |
Collapse
|
8
|
Colston JM, Chernyavskiy P, Gardner L, Nong M, Fang B, Houpt E, Swarup S, Badr H, Zaitchik B, Lakshmi V, Kosek M. The Planetary Child Health & Enterics Observatory (Plan-EO): a protocol for an interdisciplinary research initiative and web-based dashboard for mapping enteric infectious diseases and their risk factors and interventions in LMICs. RESEARCH SQUARE 2024:rs.3.rs-2640564. [PMID: 36993232 PMCID: PMC10055683 DOI: 10.21203/rs.3.rs-2640564/v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Diarrhea remains a leading cause of childhood illness throughout the world that is increasing due to climate change and is caused by various species of ecologically sensitive pathogens. The emerging Planetary Health movement emphasizes the interdependence of human health with natural systems, and much of its focus has been on infectious diseases and their interactions with environmental and human processes. Meanwhile, the era of big data has engendered a public appetite for interactive web-based dashboards for infectious diseases. However, enteric infectious diseases have been largely overlooked by these developments. Methods The Planetary Child Health and Enterics Observatory (Plan-EO) is a new initiative that builds on existing partnerships between epidemiologists, climatologists, bioinformaticians, and hydrologists as well as investigators in numerous low- and middle-income countries. Its objective is to provide the research and stakeholder community with an evidence base for the geographical targeting of enteropathogen-specific child health interventions such as novel vaccines. The initiative will produce, curate, and disseminate spatial data products relating to the distribution of enteric pathogens and their environmental and sociodemographic determinants. Discussion As climate change accelerates there is an urgent need for etiology-specific estimates of diarrheal disease burden at high spatiotemporal resolution. Plan-EO aims to address key challenges and knowledge gaps by making rigorously obtained, generalizable disease burden estimates freely available and accessible to the research and stakeholder communities. Pre-processed environmental and EO-derived spatial data products will be housed, continually updated, and made publicly available to the research and stakeholder communities both within the webpage itself and for download. These inputs can then be used to identify and target priority populations living in transmission hotspots and for decision-making, scenario-planning, and disease burden projection. Study registration PROSPERO protocol #CRD42023384709.
Collapse
Affiliation(s)
| | | | | | - Malena Nong
- University of Virginia College of Arts & Sciences
| | | | - Eric Houpt
- University of Virginia School of Medicine
| | | | | | | | | | | |
Collapse
|
9
|
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
|
10
|
Zhao H, Ni P, Zhao Q, Liang X, Ai D, Erhardt S, Wang J, Li Y, Wang J. Identifying the serious clinical outcomes of adverse reactions to drugs by a multi-task deep learning framework. Commun Biol 2023; 6:870. [PMID: 37620651 PMCID: PMC10449791 DOI: 10.1038/s42003-023-05243-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Adverse Drug Reactions (ADRs) have a direct impact on human health. As continuous pharmacovigilance and drug monitoring prove to be costly and time-consuming, computational methods have emerged as promising alternatives. However, most existing computational methods primarily focus on predicting whether or not the drug is associated with an adverse reaction and do not consider the core issue of drug benefit-risk assessment-whether the treatment outcome is serious when adverse drug reactions occur. To this end, we categorize serious clinical outcomes caused by adverse reactions to drugs into seven distinct classes and present a deep learning framework, so-called GCAP, for predicting the seriousness of clinical outcomes of adverse reactions to drugs. GCAP has two tasks: one is to predict whether adverse reactions to drugs cause serious clinical outcomes, and the other is to infer the corresponding classes of serious clinical outcomes. Experimental results demonstrate that our method is a powerful and robust framework with high extendibility. GCAP can serve as a useful tool to successfully address the challenge of predicting the seriousness of clinical outcomes stemming from adverse reactions to drugs.
Collapse
Affiliation(s)
- Haochen Zhao
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China
- Xiangjiang Laboratory, Changsha, 410205, China
| | - Peng Ni
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China
| | - Qichang Zhao
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China
| | - Xiao Liang
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China
| | - Di Ai
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Shannon Erhardt
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yaohang Li
- Department of Computer Science, Old Dominion University, Norfolk, VA, 23529-0001, USA
| | - Jianxin Wang
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China.
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China.
- Xiangjiang Laboratory, Changsha, 410205, China.
| |
Collapse
|
11
|
Dhalaria P, Kapur S, Singh AK, Verma A, Priyadarshini P, Taneja G. Potential impact of rotavirus vaccination on reduction of childhood diarrheal disease in India: An analysis of National Family Health Survey-5. Vaccine X 2023; 14:100319. [PMID: 37275272 PMCID: PMC10239013 DOI: 10.1016/j.jvacx.2023.100319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 06/07/2023] Open
Abstract
Rotavirus is one of the leading causes of diarrhea in infants and young children worldwide. In this study, we investigated the impact of rotavirus vaccination on the prevalence of diarrheal disease among children under five years of age in India. Research on the impact of the rotavirus vaccine on reducing diarrheal disease is therefore important in contributing to the growing body of evidence on the effectiveness of this intervention in improving child health outcomes. We adopted multivariate logistic regression and propensity score matching analysis to examine the association between diarrhea and the rotavirus vaccine. The bivariate analysis finding shows that the prevalence of diarrhea was remarkably higher (9.1%) among children who had not received rotavirus and the prevalence was 7.5%, 7.5%, and 7.2% among children who received one dose, two doses, and three rotavirus doses (all) respectively. The result of multivariate logistic regression shows that children who received all three doses of the rotavirus vaccine were 16% less likely to experience diarrhea compared to those who did not receive any rotavirus vaccine. Our analysis also found that the prevalence of diarrhea decreased significantly in the years following the introduction of the vaccine. The results of this study suggest that the rotavirus vaccine has a significant impact on reducing childhood diarrheal disease in India. These results have the potential to inform policy decisions and enable healthcare professionals to concert their efforts in reducing the diarrheal disease burden and its timely prevention in children. The study will also contribute to the existing literature on the impact of rotavirus vaccination in reducing the prevalence of diarrhea among children in India.
Collapse
Affiliation(s)
- Pritu Dhalaria
- Immunization Technical Support Unit, Ministry of Health & Family Welfare, Government of India, New Delhi 110070, India
| | | | - Ajeet Kumar Singh
- Immunization Technical Support Unit, Ministry of Health & Family Welfare, Government of India, New Delhi 110070, India
| | - Ajay Verma
- Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Pretty Priyadarshini
- Immunization Technical Support Unit, Ministry of Health & Family Welfare, Government of India, New Delhi 110070, India
| | - Gunjan Taneja
- Bill & Melinda Gates Foundation, New Delhi 110067, India
| |
Collapse
|
12
|
Sifuna P, Shaw AV, Lucas T, Ogutu B, Otieno W, Larsen DA. Deployment of Rotavirus Vaccine in Western Kenya Coincides with a Reduction in All-Cause Child Mortality: A Retrospective Cohort Study. Vaccines (Basel) 2023; 11:1299. [PMID: 37631867 PMCID: PMC10458991 DOI: 10.3390/vaccines11081299] [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: 06/22/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 08/27/2023] Open
Abstract
Rotavirus is an important cause of fatal pediatric diarrhea worldwide. Many national immunization programs began adding rotavirus vaccine following a 2009 World Health Organization recommendation. Kenya added rotavirus vaccine to their immunization program at the end of 2014. From a cohort of 38,463 children in the Kisumu health and demographic surveillance site in western Kenya, we assessed how the implementation of the rotavirus vaccine affected mortality in children under 3 years of age. Following its introduction in late 2014, the span of rotavirus vaccine coverage for children increased to 75% by 2017. Receiving the rotavirus vaccine was associated with a 44% reduction in all-cause child mortality (95% confidence interval = 28-68%, p < 0.0001), but not diarrhea-specific mortality (p = 0.401). All-cause child mortality declined 2% per month following the implementation of the rotavirus vaccine (p = 0.002) among both vaccinated and unvaccinated children, but diarrhea-specific mortality was not associated with the implementation of the rotavirus vaccine independent of individual vaccine status (p = 0.125). The incidence of acute diarrhea decreased over the study period, and the introduction of the rotavirus vaccine was not associated with population-wide trends (p = 0.452). The receipt of the rotavirus vaccine was associated with a 34% reduction in the incidence of diarrhea (95% confidence interval = 24-43% reduction). These results suggest that rotavirus vaccine may have had an impact on all-cause child mortality. The analyses of diarrhea-specific mortality were limited by relatively few deaths (n = 57), as others have found a strong reduction in diarrhea-specific mortality. Selection bias may have played a part in these results-children receiving rotavirus vaccine were more likely to be fully immunized than children not receiving the rotavirus vaccine.
Collapse
Affiliation(s)
- Peter Sifuna
- Kenya Medical Research Institute (KEMRI), Kisumu 40100, Kenya; (P.S.); (T.L.); (B.O.); (W.O.)
- US Army Medical Research Directorate–Africa (USAMRD-A), Kisumu 00200, Kenya
| | - Andrea V. Shaw
- Institute for Global Health and Translational Science, SUNY Upstate Medical University, Syracuse, NY 13210, USA;
| | - Tina Lucas
- Kenya Medical Research Institute (KEMRI), Kisumu 40100, Kenya; (P.S.); (T.L.); (B.O.); (W.O.)
- US Army Medical Research Directorate–Africa (USAMRD-A), Kisumu 00200, Kenya
| | - Bernards Ogutu
- Kenya Medical Research Institute (KEMRI), Kisumu 40100, Kenya; (P.S.); (T.L.); (B.O.); (W.O.)
- US Army Medical Research Directorate–Africa (USAMRD-A), Kisumu 00200, Kenya
| | - Walter Otieno
- Kenya Medical Research Institute (KEMRI), Kisumu 40100, Kenya; (P.S.); (T.L.); (B.O.); (W.O.)
- US Army Medical Research Directorate–Africa (USAMRD-A), Kisumu 00200, Kenya
| | - David A. Larsen
- Department of Public Health, Syracuse University, Syracuse, NY 13244, USA
| |
Collapse
|
13
|
Marti SG, Gibbons L, Reidel S, Stupka J, Degiuseppe J, Argento F, Gómez JA. Rotavirus Vaccine Impact since Its Introduction in the National Immunization Program of Argentina. Infect Dis Ther 2023; 12:513-526. [PMID: 36520328 PMCID: PMC9925648 DOI: 10.1007/s40121-022-00709-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/29/2022] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Rotavirus (RV) is the most common cause of childhood diarrhea. Argentina introduced RV vaccination in the National Immunization Program in January 2015. This study evaluates the impact of RV vaccine implementation on the burden of acute diarrheal disease (ADD) and RV positive cases, and hospitalizations among children in Argentina. METHODS A counterfactual time-series analysis was performed. Data on ADD (2013-2018) and RV diarrhea (2012-2018) cases in children aged < 5 years were collected from the National Healthcare Surveillance System (clinical and laboratory data). Data on hospital discharges following ADD and RV diarrhea (2011-2017) were retrieved from the Health Statistics and Information Office. All data were classified by the age groups < 1 year, < 2 years, 2-5 years. Vaccine impact was defined as the difference between the predicted time trend (simulated using 2012-2014 data) and the actual post-vaccination data (2015-2018). RESULTS A significant reduction of 22.1% of notified ADD cases and 15.4% of hospital discharges following ADD among children < 2 years was observed in the 3 years after RV vaccine implementation. Data also showed a significant decline of 54.0% and 59.4% of notified RV cases in children < 2 and < 1 years, respectively, and a reduction of 39.3% and 40.8% in RV hospital discharges for the same age groups. CONCLUSION This study shows a significant reduction in notified ADD cases and RV cases and hospital discharges following ADD and RV cases in children < 2 years after RV vaccine introduction in Argentina in 2015.
Collapse
Affiliation(s)
| | - Luz Gibbons
- Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
| | - Sara Reidel
- Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
| | - Juan Stupka
- Laboratorio Nacional de Referencia Para Rotavirus y Norovirus, INEI-ANLIS, Dr. Carlos G. Malbran, ANLIS, Buenos Aires, Argentina
| | - Juan Degiuseppe
- Laboratorio Nacional de Referencia Para Rotavirus y Norovirus, INEI-ANLIS, Dr. Carlos G. Malbran, ANLIS, Buenos Aires, Argentina
| | - Fernando Argento
- Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
| | | |
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
|
Omatola CA, Olaniran AO. Genetic heterogeneity of group A rotaviruses: a review of the evolutionary dynamics and implication on vaccination. Expert Rev Anti Infect Ther 2022; 20:1587-1602. [PMID: 36285575 DOI: 10.1080/14787210.2022.2139239] [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: 01/12/2023]
Abstract
INTRODUCTION Human rotavirus remains a major etiology of acute gastroenteritis among under 5-year children worldwide despite the availability of oral vaccines. The genetic instability of rotavirus and the ability to form different combinations from the different G- and P-types reshapes the antigenic landscape of emerging strains which often display limited or no antigen identities with the vaccine strain. As evidence also suggests, the selection of the antigenically distinct novel or rare strains and their successful spread in the human population has raised concerns regarding undermining the effectiveness of vaccination programs. AREAS COVERED We review aspects related to current knowledge about genetic and antigenic heterogeneity of rotavirus, the mechanism of genetic diversity and evolution, and the implication of genetic change on vaccination. EXPERT OPINION Genetic changes in the segmented genome of rotavirus can alter the antigenic landscape on the virion capsid and further promote viral fitness in a fully vaccinated population. Against this background, the potential risk of the appearance of new rotavirus strains over the long term would be better predicted by a continued and increased close monitoring of the variants across the globe to identify any change associated with disease dynamics.
Collapse
Affiliation(s)
- Cornelius A Omatola
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban, Republic of South Africa
| | - Ademola O Olaniran
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban, Republic of South Africa
| |
Collapse
|
16
|
Sarkar R, Banerjee S, Halder P, Koley H, Komoto S, Chawla-Sarkar M. Suppression of classical nuclear import pathway by importazole and ivermectin inhibits rotavirus replication. J Antimicrob Chemother 2022; 77:3443-3455. [PMID: 36210599 DOI: 10.1093/jac/dkac339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Rotavirus is the foremost cause of acute gastroenteritis among infants in resource-poor countries, causing severe morbidity and mortality. The currently available rotavirus vaccines are effective in reducing severity of the disease but not the infection rates, thus antivirals as an adjunct therapy are needed to reduce the morbidity in children. Viruses rely on host cellular machinery for nearly every step of the replication cycle. Therefore, targeting host factors that are indispensable for virus replication could be a promising strategy. OBJECTIVES To assess the therapeutic potential of ivermectin and importazole against rotaviruses. METHODS Antirotaviral activity of importazole and ivermectin was measured against various rotavirus strains (RV-SA11, RV-Wa, RV-A5-13, RV-EW) in vitro and in vivo by quantifying viral protein expression by western blot, analysing viroplasm formation by confocal microscopy, and measuring virus yield by plaque assay. RESULTS Importin-β1 and Ran were found to be induced during rotavirus infection. Knocking down importin-β1 severely impaired rotavirus replication, suggesting a critical role for importin-β1 in the rotavirus life cycle. In vitro studies revealed that treatment of ivermectin and importazole resulted in reduced synthesis of viral proteins, diminished production of infectious virus particles, and decrease in viroplasm-positive cells. Mechanistic study proved that both drugs perform antirotavirus activity by inhibiting the function of importin-β1. In vivo investigations in mice also confirmed the antirotavirus potential of importazole and ivermectin at non-toxic doses. Treatments of rotavirus-infected mice with either drug resulted in diminished shedding of viral particles in the stool sample, reduced expression of viral protein in the small intestine and restoration of damaged intestinal villi comapared to untreated infected mice. CONCLUSIONS The study highlights the potential of importazole and ivermectin as antirotavirus therapeutics.
Collapse
Affiliation(s)
- Rakesh Sarkar
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme-XM, Beliaghata, 700010, Kolkata, West Bengal, India
| | - Shreya Banerjee
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme-XM, Beliaghata, 700010, Kolkata, West Bengal, India
| | - Prolay Halder
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Satoshi Komoto
- Department of Virology and Parasitology, Fujita Health University School of Medicine, Aichi, Japan
| | - Mamta Chawla-Sarkar
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme-XM, Beliaghata, 700010, Kolkata, West Bengal, India
| |
Collapse
|
17
|
Asare EO, Hergott D, Seiler J, Morgan B, Archer H, Wiyeh AB, Guo B, Driver M, Giersing B, Hasso-Agopsowicz M, Lingappa J, Lopman BA, Pitzer VE. Case fatality risk of diarrhoeal pathogens: a systematic review and meta-analysis. Int J Epidemiol 2022; 51:1469-1480. [PMID: 35578827 PMCID: PMC9557849 DOI: 10.1093/ije/dyac098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 05/06/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Estimates of the relative contribution of different pathogens to all-cause diarrhoea mortality are needed to inform global diarrhoea burden models and prioritize interventions. We aimed to investigate and estimate heterogeneity in the case fatality risk (CFR) of different diarrhoeal pathogens. METHODS We conducted a systematic review and meta-analysis of studies that reported cases and deaths for 15 enteric pathogens published between 1990 and 2019. The primary outcome was the pathogen-specific CFR stratified by age group, country-specific under-5 mortality rate, setting, study year and rotavirus vaccine introduction status. We developed fixed-effects and multilevel mixed-effects logistic regression models to estimate the pooled CFR overall and for each pathogen, controlling for potential predictors of heterogeneity. RESULTS A total of 416 studies met review criteria and were included in the analysis. The overall crude CFR for all pathogens was 0.65%, but there was considerable heterogeneity between and within studies. The overall CFR estimated from a random-effects model was 0.04% (95% CI: 0.026%-0.062%), whereas the pathogen-specific CFR estimates ranged from 0% to 2.7%. When pathogens were included as predictors of the CFR in the overall model, the highest and lowest odds ratios were found for enteropathogenic Escherichia coli (EPEC) [odds ratio (OR) = 3.0, 95% CI: 1.28-7.07] and rotavirus (OR = 0.23, 95% CI: 0.13-0.39), respectively. CONCLUSION We provide comprehensive estimates of the CFR across different diarrhoeal pathogens and highlight pathogens for which more studies are needed. The results motivate the need for diarrhoeal interventions and could help prioritize pathogens for vaccine development.
Collapse
Affiliation(s)
- Ernest O Asare
- Corresponding author. Department of Epidemiology of Microbial Diseases, Yale School of Public Health, PO Box 208034, 60 College Street, New Haven, CT 06520-8034, USA. E-mail: ;
| | - Dianna Hergott
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Jessica Seiler
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Brooks Morgan
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Helena Archer
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Alison B Wiyeh
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Boya Guo
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Matt Driver
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Birgitte Giersing
- Vaccine Product Delivery Research, Immunization, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland
| | - Mateusz Hasso-Agopsowicz
- Vaccine Product Delivery Research, Immunization, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland
| | - Jairam Lingappa
- Departments of Global Health, Medicine, and Pediatrics, University of Washington, Seattle, WA, USA
| | - Benjamin A Lopman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Virginia E Pitzer
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT, USA
| |
Collapse
|
18
|
Lahiri KR, Singh R, Apte M, Patil M, Taksande A, Varona R, Chatterjee G, Verma M, Brette S, Perez MI. Efficacy and safety of Bacillus clausii (O/C, N/R, SIN, T) probiotic combined with oral rehydration therapy (ORT) and zinc in acute diarrhea in children: a randomized, double-blind, placebo-controlled study in India. Trop Dis Travel Med Vaccines 2022; 8:9. [PMID: 35397572 PMCID: PMC8994895 DOI: 10.1186/s40794-022-00166-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/13/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Childhood diarrhea remains a major disease burden, particularly in developing countries, and is a leading cause of death in children aged < 5 years, worldwide. Treatment of acute diarrhea now includes probiotics to potentially reduce the duration and severity of the illness. This phase 3, randomized, placebo-controlled, double-blind study assessed the efficacy and safety of four strains (O/C, N/R, SIN, T) of Bacillus clausii probiotic (Enterogermina®) plus oral rehydration therapy (ORT) and zinc, versus placebo plus ORT and zinc, in infants and children in India with acute moderate diarrhea. METHODS Patients aged 6 months to 5 years with acute moderate diarrhea (WHO 2005 definition) of < 48 h' duration were randomly assigned to receive one mini bottle of either polyantibiotic-resistant B. clausii (oral suspension of 2 billion spores per 5 mL bottle) or matching placebo twice daily (morning and evening) for 5 days. Exclusion criteria included known hypersensitivity to B. clausii or excipients in the study treatment, or to other probiotics. Patients were admitted to hospital from Day 1 and discharged ≥6 h after diarrhea resolution, or a maximum of 5 days. The primary endpoint was duration of acute diarrhea from randomization to recovery. Secondary endpoints included frequency of stools, diapers with stools, or dehydration status. RESULTS In total, 457 patients were randomized; 454 were treated. Similar proportions of patients showed recovery from diarrhea over the 120 h after randomization (97.0% in the B. clausii group [n = 227]; 98.0% on placebo [n = 227]). Median time to recovery was also similar: 42.83 (95% CI: 40.90-44.90) hours for B. clausii and 42.13 (95% CI: 39.80-43.87) hours for placebo. However, no statistically significant difference was observed between groups (hazard ratio = 0.93 [95% CI: 0.77-1.13]; p = 0.6968); nor were there statistically significant differences between groups for the secondary endpoints. Treatment with B. clausii was well tolerated with incidence of adverse events (9.7%) similar to that for placebo (12.3%). CONCLUSIONS No significant difference in efficacy between B. clausii and placebo was demonstrated. Sample size may have been inadequate to allow detection of a between-group difference in efficacy, given the mild/moderate severity (only ~ 20% of patients had nausea/vomiting or abdominal pains) and short duration of disease among subjects, the relatively late start of treatment (most were already on Day 2 of their disease episode when study treatment started) and the effectiveness of the standard of care with ORT and zinc in both treatment groups. TRIAL REGISTRATION CTRI number CTRI/2018/10/016053 . Registered on 17 October 2018. EudraCT number 2016-005165-31 . Registered 14 May 2020 (retrospectively registered).
Collapse
Affiliation(s)
- Keya Rani Lahiri
- D Y Patil Medical College and Hospital, Nerul, Navi Mumbai, India
| | - Raghvendra Singh
- Maulana Azad Medical College and Lok Nayak Hospital, 2 Bahadur Shah Zafar Road, New Delhi, India
| | - Mohini Apte
- Government Medical College and Hospital, Medical College Square, Nagpur, India
| | - Mahantesh Patil
- JN Medical College & KLES Dr. Prabhakar Kore Hospital & MRC, Nehru Nagar, Belgaum, India
| | - Amar Taksande
- Acharya Vinoba Bhave Rural Hospital, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences, Sawangi (Meghe), Wardha, India
| | - Rafael Varona
- Global Medical, Consumer Healthcare, Sanofi-Aventis, Gentilly, France
| | - Godhuli Chatterjee
- Senior Medical Advisor and Clinical Study Unit Safety Lead, Clinical Study Unit (India-South East Asia Cluster), Sanofi Healthcare India Private Limited, Mumbai, Maharashtra, India
| | - Manish Verma
- Sanofi Healthcare India Private Limited, Mumbai, Maharashtra, India
| | | | - Marcos Iii Perez
- Global Medical, Consumer Healthcare, Sanofi-Aventis Deutschland GmbH, Frankfurt, Germany.
| |
Collapse
|
19
|
Mado S, Giwa F, Abdullahi S, Alfa A, Yaqub Y, Usman Y, Wammanda R, Mwenda J, Isiaka A, Yusuf K, Lawali N. Prevalence and characteristics of rotavirus acute gastroenteritis among under-five children in ahmadu bello university teaching hospital, Zaria, Nigeria. Ann Afr Med 2022; 21:283-287. [PMID: 36204917 PMCID: PMC9671188 DOI: 10.4103/aam.aam_31_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background: Rotavirus infection is a significant cause of gastroenteritis in developing countries and, in severe cases even leads to death. The impact of rotavirus vaccine introduction in reducing the rotavirus disease burden in children was well known. The study was aimed to determine the prevalence and clinical characteristics of rotavirus gastroenteritis before the introduction of rotavirus vaccine into Nigeria's routine immunization program. Materials and Methods: We conducted a cross-sectional hospital-based study involving 735 children aged 0–59 months with acute gastroenteritis hospitalized at the Ahmadu Bello University Teaching Hospital Zaria from September 2017 to August 2020. Relevant sociodemographic and clinical data were obtained and entered into the World Health Organization standardized case investigation forms. Stool specimens were tested for rotavirus Group A antigen using the ProSpecT™ Rotavirus Microplate Assay by Thermoscientific Oxoid Microbiology UK. Results: One hundred and fifty-three stool samples tested positive for rotavirus giving a prevalence of 20.8%. One hundred and two (66.7%) children with rotavirus gastroenteritis were infants. There were 87 males and 66 females with M: F ratio of 1.3:1. Only 30 (19.6%) children with rotavirus-associated diarrhea presented with severe dehydration. The presence of vomiting was significantly associated with rotavirus diarrhea (P = 0.001). More cases of rotavirus diarrhea occurred in September through February. None of the studied children were vaccinated against rotavirus. Conclusion: The prevalence of rotavirus diarrhea remains high in this study. Infants were recognized as a high-risk group, and none of them were vaccinated against rotavirus and this underscores the urgent need for implementing the rotavirus vaccine in the national vaccination program to reduce the disease burden in the country.
Collapse
|
20
|
Mandolo JJ, Henrion MYR, Mhango C, Chinyama E, Wachepa R, Kanjerwa O, Malamba-Banda C, Shawa IT, Hungerford D, Kamng’ona AW, Iturriza-Gomara M, Cunliffe NA, Jere KC. Reduction in Severity of All-Cause Gastroenteritis Requiring Hospitalisation in Children Vaccinated against Rotavirus in Malawi. Viruses 2021; 13:2491. [PMID: 34960760 PMCID: PMC8707889 DOI: 10.3390/v13122491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 01/21/2023] Open
Abstract
Rotavirus is the major cause of severe gastroenteritis in children aged <5 years. Introduction of the G1P[8] Rotarix® rotavirus vaccine in Malawi in 2012 has reduced rotavirus-associated hospitalisations and diarrhoeal mortality. However, the impact of rotavirus vaccine on the severity of gastroenteritis presented in children requiring hospitalisation remains unknown. We conducted a hospital-based surveillance study to assess the impact of Rotarix® vaccination on the severity of gastroenteritis presented by Malawian children. Stool samples were collected from children aged <5 years who required hospitalisation with acute gastroenteritis from December 2011 to October 2019. Gastroenteritis severity was determined using Ruuska and Vesikari scores. Rotavirus was detected using enzyme immunoassay. Rotavirus genotypes were determined using nested RT-PCR. Associations between Rotarix® vaccination and gastroenteritis severity were investigated using adjusted linear regression. In total, 3159 children were enrolled. After adjusting for mid-upper arm circumference (MUAC), age, gender and receipt of other vaccines, all-cause gastroenteritis severity scores were 2.21 units lower (p < 0.001) among Rotarix®-vaccinated (n = 2224) compared to Rotarix®-unvaccinated children (n = 935). The reduction in severity score was observed against every rotavirus genotype, although the magnitude was smaller among those infected with G12P[6] compared to the remaining genotypes (p = 0.011). Each one-year increment in age was associated with a decrease of 0.43 severity score (p < 0.001). Our findings provide additional evidence on the impact of Rotarix® in Malawi, lending further support to Malawi's Rotarix® programme.
Collapse
Affiliation(s)
- Jonathan J. Mandolo
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Department of Biomedical Sciences, School of Life Sciences and Health Professions, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Marc Y. R. Henrion
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
| | - Chimwemwe Mhango
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Department of Biomedical Sciences, School of Life Sciences and Health Professions, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - End Chinyama
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
| | - Richard Wachepa
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
| | - Oscar Kanjerwa
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
| | - Chikondi Malamba-Banda
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- Department of Medical Laboratory Sciences, School of Life Sciences and Health Professions, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Isaac T. Shawa
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Department of Medical Laboratory Sciences, School of Life Sciences and Health Professions, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Daniel Hungerford
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool L69 7BE, UK
| | - Arox W. Kamng’ona
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Department of Biomedical Sciences, School of Life Sciences and Health Professions, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
| | - Miren Iturriza-Gomara
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool L69 7BE, UK
- Centre for Vaccine Innovation and Access, Program for Appropriate Technology in Health (PATH), 1218 Geneva, Switzerland
| | - Nigel A. Cunliffe
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool L69 7BE, UK
| | - Khuzwayo C. Jere
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- Department of Medical Laboratory Sciences, School of Life Sciences and Health Professions, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool L69 7BE, UK
| |
Collapse
|
21
|
Bergman H, Henschke N, Hungerford D, Pitan F, Ndwandwe D, Cunliffe N, Soares-Weiser K. Vaccines for preventing rotavirus diarrhoea: vaccines in use. Cochrane Database Syst Rev 2021; 11:CD008521. [PMID: 34788488 PMCID: PMC8597890 DOI: 10.1002/14651858.cd008521.pub6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Rotavirus is a common cause of diarrhoea, diarrhoea-related hospital admissions, and diarrhoea-related deaths worldwide. Rotavirus vaccines prequalified by the World Health Organization (WHO) include Rotarix (GlaxoSmithKline), RotaTeq (Merck), and, more recently, Rotasiil (Serum Institute of India Ltd.), and Rotavac (Bharat Biotech Ltd.). OBJECTIVES To evaluate rotavirus vaccines prequalified by the WHO for their efficacy and safety in children. SEARCH METHODS On 30 November 2020, we searched PubMed, the Cochrane Infectious Diseases Group Specialized Register, CENTRAL (published in the Cochrane Library), Embase, LILACS, Science Citation Index Expanded, Social Sciences Citation Index, Conference Proceedings Citation Index-Science, Conference Proceedings Citation Index-Social Science & Humanities. We also searched the WHO ICTRP, ClinicalTrials.gov, clinical trial reports from manufacturers' websites, and reference lists of included studies, and relevant systematic reviews. SELECTION CRITERIA We selected randomized controlled trials (RCTs) conducted in children that compared rotavirus vaccines prequalified for use by the WHO with either placebo or no intervention. DATA COLLECTION AND ANALYSIS Two authors independently assessed trial eligibility and assessed risk of bias. One author extracted data and a second author cross-checked them. We combined dichotomous data using the risk ratio (RR) and 95% confidence interval (CI). We stratified the analyses by under-five country mortality rate and used GRADE to evaluate evidence certainty. MAIN RESULTS Sixty trials met the inclusion criteria and enrolled a total of 228,233 participants. Thirty-six trials (119,114 participants) assessed Rotarix, 15 trials RotaTeq (88,934 participants), five trials Rotasiil (11,753 participants), and four trials Rotavac (8432 participants). Rotarix Infants vaccinated and followed up for the first year of life In low-mortality countries, Rotarix prevented 93% of severe rotavirus diarrhoea cases (14,976 participants, 4 trials; high-certainty evidence), and 52% of severe all-cause diarrhoea cases (3874 participants, 1 trial; moderate-certainty evidence). In medium-mortality countries, Rotarix prevented 79% of severe rotavirus diarrhoea cases (31,671 participants, 4 trials; high-certainty evidence), and 36% of severe all-cause diarrhoea cases (26,479 participants, 2 trials; high-certainty evidence). In high-mortality countries, Rotarix prevented 58% of severe rotavirus diarrhoea cases (15,882 participants, 4 trials; high-certainty evidence), and 27% of severe all-cause diarrhoea cases (5639 participants, 2 trials; high-certainty evidence). Children vaccinated and followed up for two years In low-mortality countries, Rotarix prevented 90% of severe rotavirus diarrhoea cases (18,145 participants, 6 trials; high-certainty evidence), and 51% of severe all-cause diarrhoea episodes (6269 participants, 2 trials; moderate-certainty evidence). In medium-mortality countries, Rotarix prevented 77% of severe rotavirus diarrhoea cases (28,834 participants, 3 trials; high-certainty evidence), and 26% of severe all-cause diarrhoea cases (23,317 participants, 2 trials; moderate-certainty evidence). In high-mortality countries, Rotarix prevented 35% of severe rotavirus diarrhoea cases (13,768 participants, 2 trials; moderate-certainty evidence), and 17% of severe all-cause diarrhoea cases (2764 participants, 1 trial; high-certainty evidence). RotaTeq Infants vaccinated and followed up for the first year of life In low-mortality countries, RotaTeq prevented 97% of severe rotavirus diarrhoea cases (5442 participants, 2 trials; high-certainty evidence). In medium-mortality countries, RotaTeq prevented 79% of severe rotavirus diarrhoea cases (3863 participants, 1 trial; low-certainty evidence). In high-mortality countries, RotaTeq prevented 57% of severe rotavirus diarrhoea cases (6775 participants, 2 trials; high-certainty evidence), but there is probably little or no difference between vaccine and placebo for severe all-cause diarrhoea (1 trial, 4085 participants; moderate-certainty evidence). Children vaccinated and followed up for two years In low-mortality countries, RotaTeq prevented 96% of severe rotavirus diarrhoea cases (5442 participants, 2 trials; high-certainty evidence). In medium-mortality countries, RotaTeq prevented 79% of severe rotavirus diarrhoea cases (3863 participants, 1 trial; low-certainty evidence). In high-mortality countries, RotaTeq prevented 44% of severe rotavirus diarrhoea cases (6744 participants, 2 trials; high-certainty evidence), and 15% of severe all-cause diarrhoea cases (5977 participants, 2 trials; high-certainty evidence). We did not identify RotaTeq studies reporting on severe all-cause diarrhoea in low- or medium-mortality countries. Rotasiil Rotasiil has not been assessed in any RCT in countries with low or medium child mortality. Infants vaccinated and followed up for the first year of life In high-mortality countries, Rotasiil prevented 48% of severe rotavirus diarrhoea cases (11,008 participants, 2 trials; high-certainty evidence), and resulted in little to no difference in severe all-cause diarrhoea cases (11,008 participants, 2 trials; high-certainty evidence). Children vaccinated and followed up for two years In high-mortality countries, Rotasiil prevented 44% of severe rotavirus diarrhoea cases (11,008 participants, 2 trials; high-certainty evidence), and resulted in little to no difference in severe all-cause diarrhoea cases (11,008 participants, 2 trials; high-certainty evidence). Rotavac Rotavac has not been assessed in any RCT in countries with low or medium child mortality. Infants vaccinated and followed up for the first year of life In high-mortality countries, Rotavac prevented 57% of severe rotavirus diarrhoea cases (6799 participants, 1 trial; moderate-certainty evidence), and 16% of severe all-cause diarrhoea cases (6799 participants, 1 trial; moderate-certainty evidence). Children vaccinated and followed up for two years In high-mortality countries, Rotavac prevented 54% of severe rotavirus diarrhoea cases (6541 participants, 1 trial; moderate-certainty evidence); no Rotavac studies have reported on severe all-cause diarrhoea at two-years follow-up. Safety No increased risk of serious adverse events (SAEs) was detected with Rotarix (103,714 participants, 31 trials; high-certainty evidence), RotaTeq (82,502 participants, 14 trials; moderate to high-certainty evidence), Rotasiil (11,646 participants, 3 trials; high-certainty evidence), or Rotavac (8210 participants, 3 trials; moderate-certainty evidence). Deaths were infrequent and the analysis had insufficient evidence to show an effect on all-cause mortality. Intussusception was rare. AUTHORS' CONCLUSIONS: Rotarix, RotaTeq, Rotasiil, and Rotavac prevent episodes of rotavirus diarrhoea. The relative effect estimate is smaller in high-mortality than in low-mortality countries, but more episodes are prevented in high-mortality settings as the baseline risk is higher. In high-mortality countries some results suggest lower efficacy in the second year. We found no increased risk of serious adverse events, including intussusception, from any of the prequalified rotavirus vaccines.
Collapse
Affiliation(s)
| | | | - Daniel Hungerford
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool, UK
| | | | - Duduzile Ndwandwe
- Cochrane South Africa, South African Medical Research Council , Cape Town, South Africa
| | - Nigel Cunliffe
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool, UK
| | | |
Collapse
|
22
|
Abdulrazzaq S, Jaafar FA, Mohammed ZA. Lactose versus Lactose Free Regimen in Children with Acute Diarrhea. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Acute diarrhea (AD) is the most frequent gastroenterological disorder, and the main cause of dehydration in childhood, and it is one of the most important causes of morbidity and mortality in children.
AIM: Assessment of lactose-free formula effect in nutritional treatment in formula-fed children with acute diarrhea.
PATIENTS AND METHODS: A cross-section interventional study carried out on 60 formula-fed children, under two years, referring with acute diarrhea, 30 children obtain lactose-free formulation and 30 children with no lactose-containing formula. According to the period of diarrheal stop and weight, changes compering two groups.
RESULTS: Thirty-two males and 28 females children (7.25 ± 5.1 months) included. Children with lactose-free formula had a significantly slighter dated to diarrhea relief compared with control group children (p < 0.01). No variance between two groups in weight change (6.9 ± 3.03 vs. 7.05 ± 3.07 kg, p = 0.2). (46.6%) of patients on Lactose free formula were discharged on the third post admission day, lactose-free formula has more effective recovery to those on cow milk formula (4.1 ± 1.2 vs. 6 ± 1.3 days, p < 0.01 significant).
CONCLUSION: Quick giving of lactose-free formulation to children on formula feeding show rapid relief of acute diarrhea.
Collapse
|
23
|
Re-evaluation of population-level protection conferred by a rotavirus vaccine using the 'fried-egg' approach in a rural setting in Bangladesh. Vaccine 2021; 39:5876-5882. [PMID: 34454788 PMCID: PMC8494114 DOI: 10.1016/j.vaccine.2021.08.048] [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: 04/15/2021] [Revised: 06/29/2021] [Accepted: 08/12/2021] [Indexed: 11/24/2022]
Abstract
The “fried-egg” analytic approach was applied to a cluster randomized trial (CRT). Overall analysis failed to reveal rotavirus vaccine (RV) herd protection. Same approach unmasked herd protection of other enteric vaccines failed for RV.
Background Vaccine herd protection assessed in a cluster-randomized trial (CRT) may be masked by disease transmission into the cluster from outside. However, herd effects can be unmasked using a ‘fried-egg’ approach whereby the analysis, restricted to the innermost households of clusters, ‘yolk’, creates an insulating ‘egg-white’ periphery. This approach has been demonstrated to unmask vaccine herd protection in reanalyses of cholera and typhoid vaccine CRTs. We applied this approach to an earlier CRT in Bangladesh of rotavirus vaccine (RV) whose overall analysis had failed to detect herd protection. Herein we present the results of this analysis. Methods In the study area, infants in 142 villages were randomized to receive two doses of RV with routine EPI vaccines (RV villages) or only EPI vaccines (non-RV villages). We analyzed RV protection against acute rotavirus diarrhoea for the entire cluster (P100) and P75, P50, P25 clusters, representing 75%, 50% and 25% of the innermost households for each cluster, respectively. Results During 2 years of follow-up, there was evidence of 27% overall (95 %CI: 7, 43) and 42% total protection (95 %CI: 23, 56) in the P100 cluster, but it did not increase when moved in smaller yolks. There was no evidence of indirect vaccine protection in the yolks at any cluster size. Conclusion Our reanalysis of the CRT using the fried- egg approach did not detect RV herd protection. Whether these findings reflect a true inability of the RV to confer herd protection in this setting, or are due to limitations of the approach, requires further study.
Collapse
|
24
|
Abstract
Rotavirus is a major cause of severe pediatric diarrhea worldwide. In 2006, two live, oral rotavirus vaccines, Rotarix and RotaTeq, were licensed for use in infants and were rapidly adopted in many high- and middle-income settings where efficacy had been demonstrated in clinical trials. Following completion of additional successful trials in low-income settings, the World Health Organization (WHO) recommended rotavirus vaccination for all infants globally in 2009. In 2018, two new rotavirus vaccines, Rotasiil and Rotavac, were prequalified by WHO, further expanding global availability. As of March 2021, rotavirus vaccines have been introduced nationally in 106 countries. Since introduction, rotavirus vaccines have demonstrated effectiveness against severe disease and mortality, even among age groups not eligible for vaccination. Cross-genotypic protection has also been demonstrated, and the favorable benefit-risk profile of these vaccines continues to be confirmed via post-marketing surveillance. Ongoing research seeks to better understand reasons for the lower effectiveness observed in lower-resource settings, and to use these findings to optimize vaccine strategies worldwide.
Collapse
Affiliation(s)
- Rachel M Burke
- Viral Gastroenteritis Branch, U.S. Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jacqueline E Tate
- Viral Gastroenteritis Branch, U.S. Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Umesh D Parashar
- Viral Gastroenteritis Branch, U.S. Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
25
|
Clinical and molecular epidemiological characterization of rotavirus infections in children under five years old in Shandong province, China. Arch Virol 2021; 166:2479-2486. [PMID: 34218319 DOI: 10.1007/s00705-021-05161-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/17/2021] [Indexed: 10/20/2022]
Abstract
Rotaviruses are important causative agents of acute gastroenteritis in children. In China, rotavirus infection has a prevalence rate of 30% and is therefore considered a serious public health problem. This study was carried out to investigate the clinical and molecular epidemiological characteristics of rotavirus infections in children under 5 years old with acute diarrhea in Shandong province, China. From July 2017 to June 2018, a total of 1211 fecal specimens were tested, and the prevalence of rotavirus infection was 32.12%. The mean age of the infected children was 12.2 ± 10.9 months, and the highest infection rate was observed in children aged 7-12 months, with a rate of 41.64%. G9P[8] (76.61%) was the most prevalent genotype combination, followed by G2P[4] (7.20%), G3P[8] (3.60%), and G9P[4] (2.06%). In addition to diarrhea, vomiting, fever, and dehydration were the most common clinical signs. In general, there was no significant difference in clinical manifestations among different age groups. However, the clinical manifestations differed significantly between vaccinated and unvaccinated children. Vaccinated children showed lower incidence and frequency of vomiting, lower incidence and degree of dehydration, and lower incidence of severe cases than unvaccinated children. These findings suggest that it is necessary to continuously monitor changes in the characteristics of rotavirus infections. Moreover, the introduction of vaccines into the national immunization program to prevent and control rotavirus infection is needed in China.
Collapse
|
26
|
Guzman-Holst A, Ortega-Barria E, Flores ÁA, Carreño-Manjarrez R, Constenla D, Cervantes-Apolinar MY. 15-year experience with rotavirus vaccination in Mexico: a systematic literature review. Hum Vaccin Immunother 2021; 17:3623-3637. [PMID: 34187326 PMCID: PMC8437458 DOI: 10.1080/21645515.2021.1936859] [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] [Indexed: 11/03/2022] Open
Abstract
A systematic review was conducted in Mexico to consolidate and evaluate evidence after 15 years of rotavirus vaccination, according to the National Immunization Program. Five databases were screened to identify published articles (January 2000-February 2020) with evidence on all clinical and epidemiological endpoints (e.g. immunogenicity, safety, efficacy, impact/effectiveness) of rotavirus vaccination in Mexico. Twenty-two articles were identified (observational studies including health-economic models: 17; randomized controlled trials: 5). Fourteen studies evaluated a human attenuated vaccine (HRV), four studies evaluated both vaccines, and only two evaluated a bovine-human reassortant vaccine, with local efficacy data only for HRV. Local evidence shows vaccines are safe, immunogenic, efficacious, and provide an acceptable risk-benefit profile. The benefits of both vaccines in alleviating the burden of all-cause diarrhea mortality and morbidity are documented in several local post-licensure studies. Findings signify overall benefits of rotavirus vaccination and support the continued use of rotavirus vaccine in Mexico.
Collapse
|
27
|
King C, Bar-Zeev N, Phiri T, Beard J, Mvula H, Crampin A, Heinsbroek E, Hungerford D, Lewycka S, Verani J, Whitney C, Costello A, Mwansambo C, Cunliffe N, Heyderman R, French N. Population impact and effectiveness of sequential 13-valent pneumococcal conjugate and monovalent rotavirus vaccine introduction on infant mortality: prospective birth cohort studies from Malawi. BMJ Glob Health 2021; 5:bmjgh-2020-002669. [PMID: 32912855 PMCID: PMC7482521 DOI: 10.1136/bmjgh-2020-002669] [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: 04/16/2020] [Revised: 06/18/2020] [Accepted: 06/24/2020] [Indexed: 11/08/2022] Open
Abstract
Background Pneumococcal conjugate vaccine (PCV) and rotavirus vaccine (RV) are key tools for reducing common causes of infant mortality. However, measurement of population-level mortality impact is lacking from sub-Saharan Africa. We evaluated mortality impact and vaccine effectiveness (VE) of PCV13 introduced in November 2011, with subsequent RV1 roll-out in October 2012, in Malawi. Methods We conducted two independent community-based birth cohort studies. Study 1, in northern Malawi (40000population), evaluated population impact using change-point analysis and negative-binomial regression of non-traumatic 14–51-week infant mortality preintroduction (1 January 2004 to 31 September 2011) and postintroduction (1 October 2011 to 1 July 2019), and against three-dose coverage. Study 2, in central Malawi (465 000 population), was recruited from 24 November 2011 to 1 June 2015. In the absence of preintroduction data, individual three-dose versus zero-dose VE was estimated using individual-level Cox survival models. In both cohorts, infants were followed with household visits to ascertain vaccination, socioeconomic and survival status. Verbal autopsies were conducted for deaths. Results Study 1 included 20 291 live births and 216 infant deaths. Mortality decreased by 28.6% (95% CI: 15.3 to 39.8) post-PCV13 introduction. A change point was identified in November 2012. Study 2 registered 50 731 live births, with 454 deaths. Infant mortality decreased from 17 to 10/1000 live births during the study period. Adjusted VE was 44.6% overall (95% CI: 23.0 to 59.1) and 48.3% (95% CI: −5.9 to 74.1) against combined acute respiratory infection, meningitis and sepsis-associated mortality. Conclusion These data provide population-level evidence of infant mortality reduction following sequential PCV13 and RV1 introduction into an established immunisation programme in Malawi. These data support increasing coverage of vaccine programmes in high-burden settings.
Collapse
Affiliation(s)
- Carina King
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
- Institute for Global Health, University College London, London, London, UK
| | - Naor Bar-Zeev
- International Vaccine Access Center, Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Centre for Global Vaccine Research, Institute of Infection & Global Health, University of Liverpool, Liverpool, Merseyside, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Tambosi Phiri
- MaiMwana Project, Parent and Child Health Initiative, Lilongwe, Malawi
| | - James Beard
- Institute for Global Health, University College London, London, London, UK
| | - Hazzie Mvula
- Karonga Prevention Study, Malawi Epidemiology and Intervention Research Unit, Chilumba, Malawi
| | - Amelia Crampin
- Karonga Prevention Study, Malawi Epidemiology and Intervention Research Unit, Chilumba, Malawi
- Institute of Health & Wellbeing, University of Glasgow, Glasgow, UK
- Department of Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Ellen Heinsbroek
- Centre for Global Vaccine Research, Institute of Infection & Global Health, University of Liverpool, Liverpool, Merseyside, UK
- Karonga Prevention Study, Malawi Epidemiology and Intervention Research Unit, Chilumba, Malawi
| | - Dan Hungerford
- Centre for Global Vaccine Research, Institute of Infection & Global Health, University of Liverpool, Liverpool, Merseyside, UK
| | - Sonia Lewycka
- Nuffield Department of Medicine, Centre for Tropical Medicine, University of Oxford, Oxford, Oxfordshire, UK
| | - Jennifer Verani
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Cynthia Whitney
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Anthony Costello
- Institute for Global Health, University College London, London, London, UK
| | - Charles Mwansambo
- MaiMwana Project, Parent and Child Health Initiative, Lilongwe, Malawi
- Ministry of Health, Lilongwe, Malawi
| | - Nigel Cunliffe
- Centre for Global Vaccine Research, Institute of Infection & Global Health, University of Liverpool, Liverpool, Merseyside, UK
| | - Rob Heyderman
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
- NIHR Global Health Research Unit on Mucosal Pathogens, Division of Infection & Immunity, University College London, London, UK
| | - Neil French
- Centre for Global Vaccine Research, Institute of Infection & Global Health, University of Liverpool, Liverpool, Merseyside, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
| |
Collapse
|
28
|
Fix A, Kirkwood CD, Steele D, Flores J. Next-generation rotavirus vaccine developers meeting: Summary of a meeting sponsored by PATH and the bill & melinda gates foundation (19-20 June 2019, Geneva). Vaccine 2020; 38:8247-8254. [PMID: 33234304 DOI: 10.1016/j.vaccine.2020.11.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 10/22/2022]
Abstract
Despite the contribution of currently licensed live, oral rotavirus vaccines (LORVs) to alleviating the burden of severe disease and death from rotavirus gastroenteritis, those vaccines have proven less efficacious in resource-limited settings than in high- and middle-income countries. It has been proposed that the residual burden of rotavirus disease might be overcome with parenterally administered vaccines, or next-generation rotavirus vaccines (NGRV). To better define the progress of development of these vaccines, a meeting of vaccine developers and manufacturers engaged in NGRV research and development was convened in Geneva in June 2019. Several NRGVs are in various stages of preclinical development, and two have already entered clinical testing. The vaccine platforms include subunit protein, inactivated whole virus, virus-like particle and RNA-based vaccines. Meeting participants included groups involved in NGRV development, scientists investigating correlates of protection of rotavirus vaccines, and representatives of international organizations with insight into considerations for vaccine introduction. This report summarizes the presentations shared at the meeting.
Collapse
|
29
|
Using the Past to Maximize the Success Probability of Future Anti-Viral Vaccines. Vaccines (Basel) 2020; 8:vaccines8040566. [PMID: 33019507 PMCID: PMC7712378 DOI: 10.3390/vaccines8040566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/16/2020] [Accepted: 09/25/2020] [Indexed: 11/16/2022] Open
Abstract
Rapid obtaining of safe, effective, anti-viral vaccines has recently risen to the top of the international agenda. To maximize the success probability of future anti-viral vaccines, the anti-viral vaccines successful in the past are summarized here by virus type and vaccine type. The primary focus is on viruses with both single-stranded RNA genomes and a membrane envelope, given the pandemic past of influenza viruses and coronaviruses. The following conclusion is reached, assuming that success of future strategies is positively correlated with strategies successful in the past. The primary strategy, especially for emerging pandemic viruses, should be development of vaccine antigens that are live-attenuated viruses; the secondary strategy should be development of vaccine antigens that are inactivated virus particles. Support for this conclusion comes from the complexity of immune systems. These conclusions imply the need for a revision in current strategic planning.
Collapse
|
30
|
Gastañaduy PA, Parashar UD. Efficient transmission of viral gastroenteritis in Dutch households. THE LANCET. INFECTIOUS DISEASES 2020; 20:519-520. [PMID: 32087774 DOI: 10.1016/s1473-3099(20)30054-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 06/10/2023]
Affiliation(s)
- Paul A Gastañaduy
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA.
| | - Umesh D Parashar
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| |
Collapse
|
31
|
Parashar UD, Tate JE. The control of diarrhea, the case of a rotavirus vaccine. SALUD PUBLICA DE MEXICO 2020; 62:1-5. [PMID: 31869556 PMCID: PMC12046585 DOI: 10.21149/9943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
[No disponible]
Collapse
Affiliation(s)
- Umesh D Parashar
- Viral Gastroenteritis Branch, Division of Viral Diseases, Centers for Disease Control and Prevention. Atlanta GA, USA
| | - Jacqueline E Tate
- Viral Gastroenteritis Branch, Division of Viral Diseases, Centers for Disease Control and Prevention. Atlanta GA, USA
| |
Collapse
|
32
|
García Martí S, Augustovski F, Gibbons L, Loggia V, Lepetic A, Gómez J, Pichón Riviere A. Impact assessment of the incorporation of the rotavirus vaccine in the province of San Luis - Argentina. Epidemiol Infect 2019; 147:e308. [PMID: 31771674 PMCID: PMC7003627 DOI: 10.1017/s0950268819001936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/11/2019] [Accepted: 10/17/2019] [Indexed: 11/10/2022] Open
Abstract
Rotavirus (RV) is the main cause of acute gastroenteritis (AGE) in young children. The San Luis province of Argentina introduced RV vaccination in May 2013. We estimate vaccine impact (RVI) using real-world data. Data on all-cause AGE cases and AGE-related hospitalisations for San Luis and the adjacent Mendoza province (control group) were obtained and analysed by interrupted time-series methods. Regardless of the model used for counterfactual predictions, we estimated a reduction in the number of all-cause AGE cases of 20-25% and a reduction in AGE-related hospitalisations of 55-60%. The vaccine impact was similar for each age group considered (<1 year, <2 years and <5 years). RV vaccination was estimated to have reduced direct medical costs in the province by about 4.5 million pesos from May 2013 to December 2014. Similar to previous studies, we found a higher impact of RV vaccination in preventing severe all-cause AGE cases requiring hospitalisation than in preventing all-cases AGE cases presenting for medical care. An assessment of the economic value of RV vaccination could take other benefits into account in addition to the avoided medical costs and the costs of vaccination.
Collapse
Affiliation(s)
- S. García Martí
- Instituto de Efectividad Clínica y Sanitaria, Dr Emilio Ravignani 2024 (C1414CPV), Buenos Aires, Argentina
| | - F. Augustovski
- Instituto de Efectividad Clínica y Sanitaria, Dr Emilio Ravignani 2024 (C1414CPV), Buenos Aires, Argentina
| | - L. Gibbons
- Instituto de Efectividad Clínica y Sanitaria, Dr Emilio Ravignani 2024 (C1414CPV), Buenos Aires, Argentina
| | - V. Loggia
- GSK, Av. Carlos Casares 3690, B1644, Victoria, Buenos Aires, Argentina
| | - A. Lepetic
- GSK, Av. Carlos Casares 3690, B1644, Victoria, Buenos Aires, Argentina
| | - J.A. Gómez
- GSK, Av. Carlos Casares 3690, B1644, Victoria, Buenos Aires, Argentina
| | - A. Pichón Riviere
- Instituto de Efectividad Clínica y Sanitaria, Dr Emilio Ravignani 2024 (C1414CPV), Buenos Aires, Argentina
| |
Collapse
|
33
|
Leslie HH, Doubova SV, Pérez-Cuevas R. Assessing health system performance: effective coverage at the Mexican Institute of Social Security. Health Policy Plan 2019; 34:ii67-ii76. [DOI: 10.1093/heapol/czz105] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2019] [Indexed: 12/21/2022] Open
Abstract
Abstract
Universal health coverage is a national priority in Mexico, with active efforts to expand public healthcare system access, increase financial protection and improve quality of care. We estimated effective coverage of multiple conditions within the Mexican Institute of Social Security (IMSS), which covers 62 million individuals. We identified routinely collected performance indicators at IMSS from 2016 related to use and quality of care for conditions avertable with high-quality healthcare; where candidate indicators were available, we quantified need for service from a population-representative survey and calculated effective coverage as proportion of individuals in need who experience potential health gains. We assessed subnational inequality across 32 states, and we weighted conditions by relative contribution to national disease burden to estimate composite effective coverage. Conditions accounting for 51% of healthcare-avertable disability-adjusted life years lost in Mexico could be assessed: antenatal care, delivery care, newborn care, childhood diarrhoea, cardiovascular disease and diabetes. Estimated effective coverage ranged from a low of 27% for childhood diarrhoea to a high of 74% for newborn care. Substantial inequality in effective coverage existed between states, particularly for maternal and child conditions. Overall effective coverage of these six conditions in IMSS was 49% in 2016. Gaps in use and quality of care must be addressed to ensure good health for all in Mexico. Despite extensive monitoring of health status and services in Mexico, currently available data are inadequate to the task of fully and routinely assessing health system effective coverage. Leaders at IMSS and similar healthcare institutions must be more purposeful in planning the assessment of population need, utilization of care and quality impacts of care to enable linkage of these data and disaggregation by location or population sub-group. Only then can complex health systems be fairly and fully evaluated.
Collapse
Affiliation(s)
- Hannah H Leslie
- Department of Global Health and Population, Harvard TH Chan School of Public Health, 90 Smith Street, 3rd floor, Boston, MA, USA
| | - Svetlana V Doubova
- Epidemiology and Health Services Research Unit CMN Siglo XXI, Mexican Institute of Social Security, Cuidad de México, Av. Cuauhtémoc 330, Doctores, PC, Mexico
| | - Ricardo Pérez-Cuevas
- Health System Research Center, National Institute of Public Health, Avenida Universidad 655, Santa María Ahuacatitlán, Cuernavaca, Mexico
- Division of Social Protection and Health, Jamaica Country Office, Inter-American Development Bank, 6 Montrose Road, Kingston, Jamaica
| |
Collapse
|
34
|
Soares‐Weiser K, Bergman H, Henschke N, Pitan F, Cunliffe N. Vaccines for preventing rotavirus diarrhoea: vaccines in use. Cochrane Database Syst Rev 2019; 2019:CD008521. [PMID: 31684685 PMCID: PMC6816010 DOI: 10.1002/14651858.cd008521.pub5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Rotavirus results in more diarrhoea-related deaths in children under five years than any other single agent in countries with high childhood mortality. It is also a common cause of diarrhoea-related hospital admissions in countries with low childhood mortality. Rotavirus vaccines that have been prequalified by the World Health Organization (WHO) include a monovalent vaccine (RV1; Rotarix, GlaxoSmithKline), a pentavalent vaccine (RV5; RotaTeq, Merck), and, more recently, another monovalent vaccine (Rotavac, Bharat Biotech). OBJECTIVES To evaluate rotavirus vaccines prequalified by the WHO (RV1, RV5, and Rotavac) for their efficacy and safety in children. SEARCH METHODS On 4 April 2018 we searched MEDLINE (via PubMed), the Cochrane Infectious Diseases Group Specialized Register, CENTRAL (published in the Cochrane Library), Embase, LILACS, and BIOSIS. We also searched the WHO ICTRP, ClinicalTrials.gov, clinical trial reports from manufacturers' websites, and reference lists of included studies and relevant systematic reviews. SELECTION CRITERIA We selected randomized controlled trials (RCTs) in children comparing rotavirus vaccines prequalified for use by the WHO versus placebo or no intervention. DATA COLLECTION AND ANALYSIS Two review authors independently assessed trial eligibility and assessed risks of bias. One review author extracted data and a second author cross-checked them. We combined dichotomous data using the risk ratio (RR) and 95% confidence interval (CI). We stratified the analysis by country mortality rate and used GRADE to evaluate evidence certainty. MAIN RESULTS Fifty-five trials met the inclusion criteria and enrolled a total of 216,480 participants. Thirty-six trials (119,114 participants) assessed RV1, 15 trials (88,934 participants) RV5, and four trials (8432 participants) Rotavac. RV1 Children vaccinated and followed up the first year of life In low-mortality countries, RV1 prevents 84% of severe rotavirus diarrhoea cases (RR 0.16, 95% CI 0.09 to 0.26; 43,779 participants, 7 trials; high-certainty evidence), and probably prevents 41% of cases of severe all-cause diarrhoea (RR 0.59, 95% CI 0.47 to 0.74; 28,051 participants, 3 trials; moderate-certainty evidence). In high-mortality countries, RV1 prevents 63% of severe rotavirus diarrhoea cases (RR 0.37, 95% CI 0.23 to 0.60; 6114 participants, 3 trials; high-certainty evidence), and 27% of severe all-cause diarrhoea cases (RR 0.73, 95% CI 0.56 to 0.95; 5639 participants, 2 trials; high-certainty evidence). Children vaccinated and followed up for two years In low-mortality countries, RV1 prevents 82% of severe rotavirus diarrhoea cases (RR 0.18, 95% CI 0.14 to 0.23; 36,002 participants, 9 trials; high-certainty evidence), and probably prevents 37% of severe all-cause diarrhoea episodes (rate ratio 0.63, 95% CI 0.56 to 0.71; 39,091 participants, 2 trials; moderate-certainty evidence). In high-mortality countries RV1 probably prevents 35% of severe rotavirus diarrhoea cases (RR 0.65, 95% CI 0.51 to 0.83; 13,768 participants, 2 trials; high-certainty evidence), and 17% of severe all-cause diarrhoea cases (RR 0.83, 95% CI 0.72 to 0.96; 2764 participants, 1 trial; moderate-certainty evidence). No increased risk of serious adverse events (SAE) was detected (RR 0.88 95% CI 0.83 to 0.93; high-certainty evidence). There were 30 cases of intussusception reported in 53,032 children after RV1 vaccination and 28 cases in 44,214 children after placebo or no intervention (RR 0.70, 95% CI 0.46 to 1.05; low-certainty evidence). RV5 Children vaccinated and followed up the first year of life In low-mortality countries, RV5 probably prevents 92% of severe rotavirus diarrhoea cases (RR 0.08, 95% CI 0.03 to 0.22; 4132 participants, 5 trials; moderate-certainty evidence). We did not identify studies reporting on severe all-cause diarrhoea in low-mortality countries. In high-mortality countries, RV5 prevents 57% of severe rotavirus diarrhoea (RR 0.43, 95% CI 0.29 to 0.62; 5916 participants, 2 trials; high-certainty evidence), but there is probably little or no difference between vaccine and placebo for severe all-cause diarrhoea (RR 0.80, 95% CI 0.58 to 1.11; 1 trial, 4085 participants; moderate-certainty evidence). Children vaccinated and followed up for two years In low-mortality countries, RV5 prevents 82% of severe rotavirus diarrhoea cases (RR 0.18, 95% CI 0.08 to 0.39; 7318 participants, 4 trials; moderate-certainty evidence). We did not identify studies reporting on severe all-cause diarrhoea in low-mortality countries. In high-mortality countries, RV5 prevents 41% of severe rotavirus diarrhoea cases (RR 0.59, 95% CI 0.43 to 0.82; 5885 participants, 2 trials; high-certainty evidence), and 15% of severe all-cause diarrhoea cases (RR 0.85, 95% CI 0.75 to 0.98; 5977 participants, 2 trials; high-certainty evidence). No increased risk of serious adverse events (SAE) was detected (RR 0.93 95% CI 0.86 to 1.01; moderate to high-certainty evidence). There were 16 cases of intussusception in 43,629 children after RV5 vaccination and 20 cases in 41,866 children after placebo (RR 0.77, 95% CI 0.41 to 1.45; low-certainty evidence). Rotavac Children vaccinated and followed up the first year of life Rotavac has not been assessed in any RCT in countries with low child mortality. In India, a high-mortality country, Rotavac probably prevents 57% of severe rotavirus diarrhoea cases (RR 0.43, 95% CI 0.30 to 0.60; 6799 participants, moderate-certainty evidence); the trial did not report on severe all-cause diarrhoea at one-year follow-up. Children vaccinated and followed up for two years Rotavac probably prevents 54% of severe rotavirus diarrhoea cases in India (RR 0.46, 95% CI 0.35 to 0.60; 6541 participants, 1 trial; moderate-certainty evidence), and 16% of severe all-cause diarrhoea cases (RR 0.84, 95% CI 0.71 to 0.98; 6799 participants, 1 trial; moderate-certainty evidence). No increased risk of serious adverse events (SAE) was detected (RR 0.93 95% CI 0.85 to 1.02; moderate-certainty evidence). There were eight cases of intussusception in 5764 children after Rotavac vaccination and three cases in 2818 children after placebo (RR 1.33, 95% CI 0.35 to 5.02; very low-certainty evidence). There was insufficient evidence of an effect on mortality from any rotavirus vaccine (198,381 participants, 44 trials; low- to very low-certainty evidence), as the trials were not powered to detect an effect at this endpoint. AUTHORS' CONCLUSIONS RV1, RV5, and Rotavac prevent episodes of rotavirus diarrhoea. Whilst the relative effect estimate is smaller in high-mortality than in low-mortality countries, there is a greater number of episodes prevented in these settings as the baseline risk is much higher. We found no increased risk of serious adverse events. 21 October 2019 Up to date All studies incorporated from most recent search All published trials found in the last search (4 Apr, 2018) were included and 15 ongoing studies are currently awaiting completion (see 'Characteristics of ongoing studies').
Collapse
Affiliation(s)
- Karla Soares‐Weiser
- CochraneEditorial & Methods DepartmentSt Albans House, 57 ‐ 59 HaymarketLondonUKSW1Y 4QX
| | - Hanna Bergman
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
| | - Nicholas Henschke
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
| | - Femi Pitan
- Chevron Corporation2 Chevron DriveLekkiLagosNigeria
| | - Nigel Cunliffe
- University of LiverpoolInstitute of Infection and Global Health, Faculty of Health and Life SciencesLiverpoolUKL69 7BE
| | | |
Collapse
|
35
|
Ndungo E, Pasetti MF. Functional antibodies as immunological endpoints to evaluate protective immunity against Shigella. Hum Vaccin Immunother 2019; 16:197-205. [PMID: 31287754 PMCID: PMC7670857 DOI: 10.1080/21645515.2019.1640427] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The development, clinical advancement and licensure of vaccines, and monitoring of vaccine effectiveness could be expedited and simplified by the ability to measure immunological endpoints that can predict a favorable clinical outcome. Antigen-specific and functional antibodies have been described in the context of naturally acquired immunity and vaccination against Shigella, and their presence in serum has been associated with reduced risk of disease in human subjects. The relevance of these antibodies as correlates of protective immunity, their mechanistic contribution to protection (e.g. target antigens, interference with pathogenesis, and participation in microbial clearance), and factors that influence their magnitude and makeup (e.g. host age, health condition, and environment) are important considerations that need to be explored. In addition to facilitating vaccine evaluation, immunological correlates of protection could be useful for identifying groups at risk and advancing immune therapies. Herein we discuss the precedent and value of functional antibodies as immunological endpoints to predict vaccine efficacy and the relevance of functional antibody activity to evaluate protective immunity against shigellosis.
Collapse
Affiliation(s)
- Esther Ndungo
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marcela F Pasetti
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
36
|
Martinez-Gutierrez M, Arcila-Quiceno V, Trejos-Suarez J, Ruiz-Saenz J. Prevalence and molecular typing of rotavirus in children with acute diarrhoea in Northeastern Colombia. Rev Inst Med Trop Sao Paulo 2019; 61:e34. [PMID: 31269110 PMCID: PMC6609135 DOI: 10.1590/s1678-9946201961034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/26/2019] [Indexed: 12/15/2022] Open
Abstract
After the introduction of the rotavirus vaccine, the number of
rotavirus-associated deaths and the predicted annual rotavirus detection rate
had slightly declined worldwide. Taking in account that in Colombia, Rotarix
vaccine was introduced in 2009, the purpose of this study was to evaluate the
presence of rotavirus A in children under five years who were treated for acute
diarrhoea in Bucaramanga, Colombia and, moreover, to determine the genotypes of
rotavirus present in those children. We performed an analytical cross-sectional
study of rotavirus A in faecal samples from children up to five years of age.
Stool samples were screened for rotavirus A using a lateral-flow
immunochromatographic assay and confirmed using a VP6 sandwich ELISA. Genotyping
of rotavirus A-positive samples was performed by PCR and sequencing of VP7 and
VP4 genes. The overall prevalence of rotavirus was 30.53% (95% confidence
interval [CI] 21.2 - 39.7). Most of the children with rotavirus (86.2%) had
received two doses of the rotavirus vaccine. G3 strains accounted for the vast
majority of cases (82.8%), followed by G12 strains (13.8%) and G3/G9
coinfections (3.4%). Among the P genotypes, P[8] was the most prevalent (69%),
followed by P[9] (31%). The most common G[P] genotype combination was G3P[8],
followed by G3P[9]. The main finding in this study was that rotavirus, in a
Colombian region, is still an important pathogen in children under five years
old, previously vaccinated. The results showed that different factors, such as
kindergarten attendance, could explain the epidemiology and transmission of
rotavirus in Bucaramanga.
Collapse
Affiliation(s)
- Marlen Martinez-Gutierrez
- Universidad Cooperativa de Colombia, Grupo de Investigación en Ciencias Animales, Bucaramanga, Colombia
| | - Victor Arcila-Quiceno
- Universidad Cooperativa de Colombia, Grupo de Investigación en Ciencias Animales, Bucaramanga, Colombia
| | - Juanita Trejos-Suarez
- Universidad de Santander, Facultad de Ciencias de la Salud, Programa de Bacteriología y Laboratorio Clínico, Grupo de Investigación en Manejo Clínico, Bucaramanga, Colombia
| | - Julian Ruiz-Saenz
- Universidad Cooperativa de Colombia, Grupo de Investigación en Ciencias Animales, Bucaramanga, Colombia
| |
Collapse
|
37
|
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]
|
38
|
Guerra Mendoza Y, Garric E, Leach A, Lievens M, Ofori-Anyinam O, Pirçon JY, Stegmann JU, Vandoolaeghe P, Otieno L, Otieno W, Owusu-Agyei S, Sacarlal J, Masoud NS, Sorgho H, Tanner M, Tinto H, Valea I, Mtoro AT, Njuguna P, Oneko M, Otieno GA, Otieno K, Gesase S, Hamel MJ, Hoffman I, Kaali S, Kamthunzi P, Kremsner P, Lanaspa M, Lell B, Lusingu J, Malabeja A, Aide P, Akoo P, Ansong D, Asante KP, Berkley JA, Adjei S, Agbenyega T, Agnandji ST, Schuerman L. Safety profile of the RTS,S/AS01 malaria vaccine in infants and children: additional data from a phase III randomized controlled trial in sub-Saharan Africa. Hum Vaccin Immunother 2019; 15:2386-2398. [PMID: 31012786 PMCID: PMC6816384 DOI: 10.1080/21645515.2019.1586040] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A phase III, double-blind, randomized, controlled trial (NCT00866619) in sub-Saharan Africa showed RTS,S/AS01 vaccine efficacy against malaria. We now present in-depth safety results from this study. 8922 children (enrolled at 5–17 months) and 6537 infants (enrolled at 6–12 weeks) were 1:1:1-randomized to receive 4 doses of RTS,S/AS01 (R3R) or non-malaria control vaccine (C3C), or 3 RTS,S/AS01 doses plus control (R3C). Aggregate safety data were reviewed by a multi-functional team. Severe malaria with Blantyre Coma Score ≤2 (cerebral malaria [CM]) and gender-specific mortality were assessed post-hoc. Serious adverse event (SAE) and fatal SAE incidences throughout the study were 24.2%–28.4% and 1.5%–2.5%, respectively across groups; 0.0%–0.3% of participants reported vaccination-related SAEs. The incidence of febrile convulsions in children was higher during the first 2–3 days post-vaccination with RTS,S/AS01 than with control vaccine, consistent with the time window of post-vaccination febrile reactions in this study (mostly the day after vaccination). A statistically significant numerical imbalance was observed for meningitis cases in children (R3R: 11, R3C: 10, C3C: 1) but not in infants. CM cases were more frequent in RTS,S/AS01-vaccinated children (R3R: 19, R3C: 24, C3C: 10) but not in infants. All-cause mortality was higher in RTS,S/AS01-vaccinated versus control girls (2.4% vs 1.3%, all ages) in our setting with low overall mortality. The observed meningitis and CM signals are considered likely chance findings, that – given their severity – warrant further evaluation in phase IV studies and WHO-led pilot implementation programs to establish the RTS,S/AS01 benefit-risk profile in real-life settings.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Seth Owusu-Agyei
- Kintampo Health Research Center , Kintampo , Ghana.,Diseases Control Department, London School of Hygiene and Tropical Medicine , London , UK
| | - Jahit Sacarlal
- Centro de Investigação em Saúde de Manhiça , Manhiça , Mozambique.,Faculdade de Medicina, Universidade Eduardo Mondlane (UEM) , Maputo , Mozambique
| | - Nahya Salim Masoud
- Muhimbili University of Health and Allied Sciences (MUHAS), Dar es Salaam and Ifakara Health Institute , Bagamoyo , Tanzania
| | - Hermann Sorgho
- Institut de Recherche en Science de la Santé , Nanoro , Burkina Faso
| | - Marcel Tanner
- Muhimbili University of Health and Allied Sciences (MUHAS), Dar es Salaam and Ifakara Health Institute , Bagamoyo , Tanzania.,Swiss Tropical and Public Health Institute , Basel , Switzerland.,Epidemiology and Medical Parasitology department, University of Basel , Basel , Switzerland
| | - Halidou Tinto
- Institut de Recherche en Science de la Santé , Nanoro , Burkina Faso
| | - Innocent Valea
- Institut de Recherche en Science de la Santé , Nanoro , Burkina Faso
| | - Ali Takadir Mtoro
- Muhimbili University of Health and Allied Sciences (MUHAS), Dar es Salaam and Ifakara Health Institute , Bagamoyo , Tanzania
| | - Patricia Njuguna
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research , Kilifi , Kenya.,Pwani University , Kilifi , Kenya.,University of Oxford , Oxford , UK
| | - Martina Oneko
- Kenya Medical Research Institute, Centre for Global Health Research , Kisumu , Kenya
| | | | - Kephas Otieno
- Kenya Medical Research Institute, Centre for Global Health Research , Kisumu , Kenya
| | - Samwel Gesase
- National Institute for Medical Research , Korogwe , Tanzania
| | - Mary J Hamel
- Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention , Atlanta , GA , USA
| | - Irving Hoffman
- University of North Carolina Project , Lilongwe , Malawi
| | - Seyram Kaali
- Kintampo Health Research Center , Kintampo , Ghana
| | | | - Peter Kremsner
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon and Institute of Tropical Medicine, University of Tübingen , Tübingen , Germany
| | - Miguel Lanaspa
- Centro de Investigação em Saúde de Manhiça , Manhiça , Mozambique.,Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona , Barcelona , Spain
| | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon and Institute of Tropical Medicine, University of Tübingen , Tübingen , Germany
| | - John Lusingu
- National Institute for Medical Research , Korogwe , Tanzania
| | | | - Pedro Aide
- Centro de Investigação em Saúde de Manhiça , Manhiça , Mozambique.,National Institute of Health, Ministry of Health , Maputo , Mozambique
| | - Pauline Akoo
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research , Kilifi , Kenya
| | - Daniel Ansong
- Kwame Nkrumah University of Science and Technology , Kumasi , Ghana
| | | | - James A Berkley
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research , Kilifi , Kenya.,University of Oxford , Oxford , UK
| | - Samuel Adjei
- Kwame Nkrumah University of Science and Technology , Kumasi , Ghana
| | - Tsiri Agbenyega
- Kwame Nkrumah University of Science and Technology , Kumasi , Ghana
| | - Selidji Todagbe Agnandji
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon and Institute of Tropical Medicine, University of Tübingen , Tübingen , Germany
| | | |
Collapse
|
39
|
Abstract
BACKGROUND Rotavirus results in more diarrhoea-related deaths in children under five years than any other single agent in countries with high childhood mortality. It is also a common cause of diarrhoea-related hospital admissions in countries with low childhood mortality. Rotavirus vaccines that have been prequalified by the World Health Organization (WHO) include a monovalent vaccine (RV1; Rotarix, GlaxoSmithKline), a pentavalent vaccine (RV5; RotaTeq, Merck), and, more recently, another monovalent vaccine (Rotavac, Bharat Biotech). OBJECTIVES To evaluate rotavirus vaccines prequalified by the WHO (RV1, RV5, and Rotavac) for their efficacy and safety in children. SEARCH METHODS On 4 April 2018 we searched MEDLINE (via PubMed), the Cochrane Infectious Diseases Group Specialized Register, CENTRAL (published in the Cochrane Library), Embase, LILACS, and BIOSIS. We also searched the WHO ICTRP, ClinicalTrials.gov, clinical trial reports from manufacturers' websites, and reference lists of included studies and relevant systematic reviews. SELECTION CRITERIA We selected randomized controlled trials (RCTs) in children comparing rotavirus vaccines prequalified for use by the WHO versus placebo or no intervention. DATA COLLECTION AND ANALYSIS Two review authors independently assessed trial eligibility and assessed risks of bias. One review author extracted data and a second author cross-checked them. We combined dichotomous data using the risk ratio (RR) and 95% confidence interval (CI). We stratified the analysis by country mortality rate and used GRADE to evaluate evidence certainty. MAIN RESULTS Fifty-five trials met the inclusion criteria and enrolled a total of 216,480 participants. Thirty-six trials (119,114 participants) assessed RV1, 15 trials (88,934 participants) RV5, and four trials (8432 participants) Rotavac.RV1 Children vaccinated and followed up the first year of life In low-mortality countries, RV1 prevents 84% of severe rotavirus diarrhoea cases (RR 0.16, 95% CI 0.09 to 0.26; 43,779 participants, 7 trials; high-certainty evidence), and probably prevents 41% of cases of severe all-cause diarrhoea (RR 0.59, 95% CI 0.47 to 0.74; 28,051 participants, 3 trials; moderate-certainty evidence). In high-mortality countries, RV1 prevents 63% of severe rotavirus diarrhoea cases (RR 0.37, 95% CI 0.23 to 0.60; 6114 participants, 3 trials; high-certainty evidence), and 27% of severe all-cause diarrhoea cases (RR 0.73, 95% CI 0.56 to 0.95; 5639 participants, 2 trials; high-certainty evidence).Children vaccinated and followed up for two yearsIn low-mortality countries, RV1 prevents 82% of severe rotavirus diarrhoea cases (RR 0.18, 95% CI 0.14 to 0.23; 36,002 participants, 9 trials; high-certainty evidence), and probably prevents 37% of severe all-cause diarrhoea episodes (rate ratio 0.63, 95% CI 0.56 to 0.71; 39,091 participants, 2 trials; moderate-certainty evidence). In high-mortality countries RV1 probably prevents 35% of severe rotavirus diarrhoea cases (RR 0.65, 95% CI 0.51 to 0.83; 13,768 participants, 2 trials; high-certainty evidence), and 17% of severe all-cause diarrhoea cases (RR 0.83, 95% CI 0.72 to 0.96; 2764 participants, 1 trial; moderate-certainty evidence).No increased risk of serious adverse events (SAE) was detected (RR 0.88 95% CI 0.83 to 0.93; high-certainty evidence). There were 30 cases of intussusception reported in 53,032 children after RV1 vaccination and 28 cases in 44,214 children after placebo or no intervention (RR 0.70, 95% CI 0.46 to 1.05; low-certainty evidence).RV5 Children vaccinated and followed up the first year of life In low-mortality countries, RV5 probably prevents 92% of severe rotavirus diarrhoea cases (RR 0.08, 95% CI 0.03 to 0.22; 4132 participants, 5 trials; moderate-certainty evidence). We did not identify studies reporting on severe all-cause diarrhoea in low-mortality countries. In high-mortality countries, RV5 prevents 57% of severe rotavirus diarrhoea (RR 0.43, 95% CI 0.29 to 0.62; 5916 participants, 2 trials; high-certainty evidence), but there is probably little or no difference between vaccine and placebo for severe all-cause diarrhoea (RR 0.80, 95% CI 0.58 to 1.11; 1 trial, 4085 participants; moderate-certainty evidence).Children vaccinated and followed up for two yearsIn low-mortality countries, RV5 prevents 82% of severe rotavirus diarrhoea cases (RR 0.18, 95% CI 0.08 to 0.39; 7318 participants, 4 trials; moderate-certainty evidence). We did not identify studies reporting on severe all-cause diarrhoea in low-mortality countries. In high-mortality countries, RV5 prevents 41% of severe rotavirus diarrhoea cases (RR 0.59, 95% CI 0.43 to 0.82; 5885 participants, 2 trials; high-certainty evidence), and 15% of severe all-cause diarrhoea cases (RR 0.85, 95% CI 0.75 to 0.98; 5977 participants, 2 trials; high-certainty evidence).No increased risk of serious adverse events (SAE) was detected (RR 0.93 95% CI 0.86 to 1.01; moderate to high-certainty evidence). There were 16 cases of intussusception in 43,629 children after RV5 vaccination and 20 cases in 41,866 children after placebo (RR 0.77, 95% CI 0.41 to 1.45; low-certainty evidence).Rotavac Children vaccinated and followed up the first year of life Rotavac has not been assessed in any RCT in countries with low child mortality. In India, a high-mortality country, Rotavac probably prevents 57% of severe rotavirus diarrhoea cases (RR 0.43, 95% CI 0.30 to 0.60; 6799 participants, moderate-certainty evidence); the trial did not report on severe all-cause diarrhoea at one-year follow-up.Children vaccinated and followed up for two yearsRotavac probably prevents 54% of severe rotavirus diarrhoea cases in India (RR 0.46, 95% CI 0.35 to 0.60; 6541 participants, 1 trial; moderate-certainty evidence), and 16% of severe all-cause diarrhoea cases (RR 0.84, 95% CI 0.71 to 0.98; 6799 participants, 1 trial; moderate-certainty evidence).No increased risk of serious adverse events (SAE) was detected (RR 0.93 95% CI 0.85 to 1.02; moderate-certainty evidence). There were eight cases of intussusception in 5764 children after Rotavac vaccination and three cases in 2818 children after placebo (RR 1.33, 95% CI 0.35 to 5.02; very low-certainty evidence).There was insufficient evidence of an effect on mortality from any rotavirus vaccine (198,381 participants, 44 trials; low- to very low-certainty evidence), as the trials were not powered to detect an effect at this endpoint. AUTHORS' CONCLUSIONS RV1, RV5, and Rotavac prevent episodes of rotavirus diarrhoea. Whilst the relative effect estimate is smaller in high-mortality than in low-mortality countries, there is a greater number of episodes prevented in these settings as the baseline risk is much higher. We found no increased risk of serious adverse events.
Collapse
Affiliation(s)
- Karla Soares‐Weiser
- CochraneEditorial & Methods DepartmentSt Albans House, 57 ‐ 59 HaymarketLondonUKSW1Y 4QX
| | - Hanna Bergman
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
| | - Nicholas Henschke
- CochraneCochrane ResponseSt Albans House57‐59 HaymarketLondonUKSW1Y 4QX
| | - Femi Pitan
- Chevron Corporation2 Chevron DriveLekkiLagosNigeria
| | - Nigel Cunliffe
- University of LiverpoolInstitute of Infection and Global Health, Faculty of Health and Life SciencesLiverpoolUKL69 7BE
| |
Collapse
|
40
|
Gillard P, Tamura T, Kuroki H, Morikawa Y, Moerman L, Parra J, Kitamura Y, Mihara K, Okamasa A. Immunogenicity and safety of the diphtheria, pertussis, tetanus and inactivated poliovirus vaccine when co-administered with the human rotavirus vaccine (Rotarix) in healthy Japanese infants: a phase IV randomized study. Hum Vaccin Immunother 2019; 15:800-808. [PMID: 30785851 PMCID: PMC6605875 DOI: 10.1080/21645515.2018.1564441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Rotavirus infections have been reported to account for 40–50% of all hospitalized acute gastroenteritis cases in young children (<5 years) in Japan. Since 2011, Rotarix containing the live attenuated human rotavirus RIX4414 strain (HRV) has been licensed in Japan for infants. Vaccination against rotavirus is optional in Japan whereas administration of diphtheria, pertussis, tetanus, and inactivated poliovirus (DPT-IPV) vaccine is part of the national routine immunization program. In this open-label, randomized, controlled, multicenter study, we evaluated the immunogenicity and safety of the DPT-IPV vaccine (Squarekids) administered concomitantly or staggered with the liquid HRV (Rotarix) vaccine in healthy Japanese infants. A total of 292 infants aged 6–12 weeks were randomly assigned to receive DPT-IPV vaccine and HRV vaccine co-administered (n = 147) or staggered (n = 145). Immune responses to DPT-IPV vaccine were evaluated by measuring the post-vaccination serum antibody titers/concentrations to each antigen at one month following the third dose of DPT-IPV vaccine. Seroprotection/seropositivity against each of the diphtheria, pertussis (pertussis toxin and filamentous hemagglutinin), tetanus, and poliovirus type 1, 2 and 3 antigens was 92.8% or higher in both groups. In terms of immunogenicity, DPT-IPV vaccine co-administered with HRV vaccine was shown to be non-inferior to DPT-IPV vaccine with a staggered administration. The safety profile was comparable in the two vaccine groups with no vaccine-related serious adverse events, no deaths and no cases of intussusception. These results support co-administration of HRV vaccine with DPT-IPV vaccine in Japan. ClinicalTrials.gov NCT02907216
Collapse
Affiliation(s)
| | - Tsuyoshi Tamura
- b Department of Pediatrics , Hashimoto Clinic , Tokyo , Japan
| | - Haruo Kuroki
- c Department of Pediatrics , Sotobo Children's Clinic , Chiba , Japan
| | | | | | - Jose Parra
- f Clinical Statistics , GSK , Wavre , Belgium
| | - Yurina Kitamura
- g Project Management, Japan Development Division , GSK , Tokyo , Japan
| | - Kazuko Mihara
- h Medicines Development Division , GSK , Tokyo , Japan
| | - Arisa Okamasa
- h Medicines Development Division , GSK , Tokyo , Japan
| |
Collapse
|
41
|
Wilson SE, Rosella LC, Wang J, Renaud A, Le Saux N, Crowcroft NS, Desai S, Harris T, Bolotin S, Gubbay J, Deeks SL. Equity and impact: Ontario's infant rotavirus immunization program five years following implementation. A population-based cohort study. Vaccine 2019; 37:2408-2414. [PMID: 30765171 DOI: 10.1016/j.vaccine.2019.01.061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/18/2019] [Accepted: 01/29/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Ontario implemented a publicly-funded rotavirus (RV) immunization program in 2011. Our objectives were to evaluate its impact on hospitalizations and emergency department (ED) visits for acute gastroenteritis (AGE) five years after implementation. METHODS We performed a population-based longitudinal retrospective cohort study to identify hospitalizations and ED visits for RV-AGE and overall AGE in all age groups using ICD-10 codes between August 1, 2005 and March 31, 2016. A negative binomial regression model that included the effect of time was used to calculate rates, rate ratios (RRs) and 95% confidence intervals (CIs) for AGE before and after the program's implementation, after adjusting for age, seasonality and secular trends. We examined the seasonality of RV-AGE hospitalizations among children under five before and after the program and explored its equity impact. RESULTS Following program implementation, RV-AGE hospitalizations and ED visits among children under five years declined by 76% (RR 0.24, 95% CI 0.20-0.28) and 68% (RR 0.32, 95% CI 0.21-0.50), respectively. In addition, hospitalizations and ED visits for overall AGE declined by 38% (RR 0.62, 95% CI 0.59-0.65) and 26% (RR 0.74, 95% CI 0.73-0.76), respectively, among children under age five. Significant reductions in both outcomes were also found across a range of age-strata. In the pre-program period, the mean monthly hospitalization rate for RV-AGE among children residing in the most marginalized neighbourhoods was 33% higher than those residing in the least marginalized (RR 1.33, 95% CI 1.17-1.52), this disparity was not evident in the program period (RR 0.95, 95% CI 0.69-1.32). We found no evidence of a seasonal shift in rotavirus pediatric hospitalizations. INTERPRETATION The introduction of routine infant rotavirus immunization has had a substantial population impact in Ontario. Our study confirms herd effects and suggests the program may have reduced previous inequities in the burden of pediatric rotavirus hospitalizations.
Collapse
Affiliation(s)
- Sarah E Wilson
- Public Health Ontario, Toronto, Ontario, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada; Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada.
| | - Laura C Rosella
- Public Health Ontario, Toronto, Ontario, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada; Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Jun Wang
- Public Health Ontario, Toronto, Ontario, Canada; Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | | | - Nicole Le Saux
- Division of Infectious Disease, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada; Department of Pediatrics, University of Ottawa, Ontario, Canada
| | - Natasha S Crowcroft
- Public Health Ontario, Toronto, Ontario, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada; Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Shalini Desai
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Tara Harris
- Public Health Ontario, Toronto, Ontario, Canada
| | - Shelly Bolotin
- Public Health Ontario, Toronto, Ontario, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan Gubbay
- Public Health Ontario, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Shelley L Deeks
- Public Health Ontario, Toronto, Ontario, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
42
|
Yu J, Lai S, Geng Q, Ye C, Zhang Z, Zheng Y, Wang L, Duan Z, Zhang J, Wu S, Parashar U, Yang W, Liao Q, Li Z. Prevalence of rotavirus and rapid changes in circulating rotavirus strains among children with acute diarrhea in China, 2009-2015. J Infect 2019; 78:66-74. [PMID: 30017609 PMCID: PMC11373190 DOI: 10.1016/j.jinf.2018.07.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/13/2018] [Accepted: 07/01/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Rotavirus is a leading cause of morbidity and mortality in young children worldwide. In China, the universal immunization of children with the rotavirus vaccine has not been introduced, and the two globally distributed vaccines (RotaTeq and Rotarix) are not licensed in the country. We aim to determine the prevalence and strain diversity of rotavirus in children with diarrhea aged ≤ five years across China. MATERIALS AND METHODS Sentinel-based surveillance of acute diarrhea was conducted at 213 participating hospitals in China from January 1, 2009, through December 31, 2015. Group A rotavirus (RVA) was tested by using enzyme-linked immunosorbent assays, and G- and P-genotype of RVA were tested by RT-PCR methods. RESULTS Of 33,616 children with diarrhea, 10,089 (30%) were positive for RVA; RVA-associated diarrhea was identified in 2247 (39.5%, n = 2247/5685) inpatients and 7842 (28.1%, n = 7842/27931) outpatients. Children living in low-middle-income regions suffered from the highest burden of rotavirus, with 40.7% of diarrhea cases attributed to rotavirus infection, followed by 31.3% in upper-middle-income and 11.2% in high-income regions. The majority of children (88.9%, n = 8976/10089) who tested positive for RVA were children aged ≤ 2 years. The seasonal peak of RVA was in the winter. Among all 2533 RVA strains genotyped, five strain combinations, G9P[8], G3P[8], G1P[8], G2P[4] and G3P[4], contributed to 71.3% (1807/2533) of the RVA-associated diarrhea cases. The predominant strain of RVA has rapidly evolved from G3P[8] and G1P[8] to G9P[8] in the recent years, with the proportion of G9P[8] having increased remarkably from 3.4% in 2009 to 60.9% in 2015. CONCLUSIONS The burden of diarrhea attributed to rotavirus is high in China, highlighting the potential value of vaccination. The rapid shift of RVA strains highlights the importance of conducting rotavirus surveillance to ensure that currently marketed vaccines provide protective efficacy against the circulating strains.
Collapse
Affiliation(s)
- Jianxing Yu
- MOH Key Laboratory of Systems Biology of Pathogens and Dr. Christophe Mérieux Laboratory, CAMS-Fondation Mérieux, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, 9 Dongdan 3rd Alley, Dongcheng District, Beijing 100730, China; Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China
| | - Shengjie Lai
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China; WorldPop Department of Geography and Environment, University of Southampton, Southampton SO17 1BJ, UK; School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, 130 Dongan Road, Shanghai 200032, China.; Flowminder Foundation, Roslagsgatan 17, SE-11355 Stockholm, Sweden
| | - Qibin Geng
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China; State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Chuchu Ye
- Research Base of Key Laboratory of Surveillance and Early-warning on Infectious Disease in China CDC, Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China; School of Public Health, Fudan University, Shanghai 200032, China
| | - Zike Zhang
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yaming Zheng
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China
| | - Liping Wang
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China
| | - Zhaojun Duan
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China
| | - Jing Zhang
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China
| | - Shuyu Wu
- Division of Global Health Protection, Center for Global Health, United States Centers for Disease Control and Prevention, Beijing 100600, China
| | - Umesh Parashar
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia 30329, USA
| | - Weizhong Yang
- Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China
| | - Qiaohong Liao
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China.
| | - Zhongjie Li
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Changbai Rd. 155#, Changping District, Beijing 102206, China.
| |
Collapse
|
43
|
Affiliation(s)
- Dani Cohen
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Khitam Muhsen
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
44
|
Luna-Casas G, Juliao P, Carreño-Manjarrez R, Castañeda-Prado A, Cervantes-Apolinar MY, Navarro-Rodriguez R, Sánchez-González G, Cortés-Alcalá R, DeAntonio R. Vaccine coverage and compliance in Mexico with the two-dose and three-dose rotavirus vaccines. Hum Vaccin Immunother 2018; 15:1251-1259. [PMID: 30380975 PMCID: PMC6783135 DOI: 10.1080/21645515.2018.1540827] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Worldwide, rotavirus infection has been a leading cause of severe diarrhea morbidity and mortality. Two rotavirus vaccines have been used in the National Immunization Program (NIP) in Mexico; two-dose Rotarix from 2006 to 2011 and three-dose RotaTeq since 2011. This study assessed coverage (receiving at least one dose or full dose series) in eligible infants, compliance (% completing dose series and % completing series on schedule) in eligible infants vaccinated with Rotarix (2010) versus RotaTeq (2012), using Mexican Social Security Institute data nationwide and by regions. In 2010, 80.7% received at least one dose of Rotarix, 75.6% received both doses and 57.0% received both doses on schedule. In 2012, 85.7% received at least one dose of RotaTeq, 61.0% received all three doses and 43.2% received all three doses on schedule. More eligible infants received all doses with Rotarix versus RotaTeq (p < 0.001). Among infants vaccinated with Rotarix versus RotaTeq, 93.7% versus 71.1% completed full series (p < 0.001), and 75.5% versus 70.9% completed full series on schedule (p = 0.105), respectively. The full series coverage and compliance decreased in all regions with RotaTeq compared with Rotarix. In conclusion, rotavirus vaccination has successfully reduced morbidity and mortality in children under 5 years in Mexico. This study found significant differences in full series coverage and compliance among infants and a higher proportion of completed scheduled at an earlier age in Mexico when comparing a two-dose vaccine in 2010 with a three-dose vaccine in 2012. Such differences might need to be taken into consideration to maximize NIP benefits, including early protection of the rotavirus vaccination program.
Collapse
|
45
|
Chavers T, De Oliveira LH, Parashar UD, Tate JE. Post-licensure experience with rotavirus vaccination in Latin America and the Caribbean: a systematic review and meta-analysis. Expert Rev Vaccines 2018; 17:1037-1051. [DOI: 10.1080/14760584.2018.1541409] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Tyler Chavers
- CDC Foundation for Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Umesh D. Parashar
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jacqueline E. Tate
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
46
|
Degiuseppe JI, Stupka JA. First assessment of all-cause acute diarrhoea and rotavirus-confirmed cases following massive vaccination in Argentina. Epidemiol Infect 2018; 146:1948-1954. [PMID: 30001762 PMCID: PMC6452988 DOI: 10.1017/s0950268818001954] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/13/2018] [Accepted: 06/24/2018] [Indexed: 11/06/2022] Open
Abstract
Argentina incorporated rotavirus massive vaccination in 2015. No specific strategy has been designed to accurately measure the impact of this recent introduction on the diarrhoeal disease burden in our country. We assessed post-vaccine introduction data (all-cause acute diarrhoea and rotavirus laboratory-confirmed cases, and genotype distribution), compared with pre-vaccination period in children under 5 years of age in Argentina. Cross-sectional ecologic analysis was conducted with data from the Argentine Surveillance Health System. Endemic channel and global and seasonal incidence rates of pre- and post-vaccination periods were calculated and further compared. Conventional binary genotypification on rotavirus-positive samples was also performed. In post-vaccination period, a global decrease of 20.8% in the rate of all-cause acute diarrhoea cases was found. The endemic channel showed that declination was more significant in the autumn/winter season. Rotavirus laboratory-confirmed cases showed 61.7% of reduction and the weekly distribution analyses indicated a significant flattening of the expected seasonal peak. G2P[4] was the most prevalent circulating genotype (57.2%). This study represents the first assessment of diarrhoeal disease burden since rotavirus massive vaccination strategy was implemented in Argentina. This introduction represented a successful intervention due to the significant decrease in all-cause acute diarrhoea cases and rotavirus laboratory-confirmed cases.
Collapse
Affiliation(s)
- J. I. Degiuseppe
- Laboratorio de Gastroenteritis Virales, Instituto Nacional de Enfermedades Infecciosas (INEI-ANLIS ‘Dr. Carlos G. Malbrán’), Argentina
| | - J. A. Stupka
- Laboratorio de Gastroenteritis Virales, Instituto Nacional de Enfermedades Infecciosas (INEI-ANLIS ‘Dr. Carlos G. Malbrán’), Argentina
| |
Collapse
|
47
|
Troeger C, Khalil IA, Rao PC, Cao S, Blacker BF, Ahmed T, Armah G, Bines JE, Brewer TG, Colombara DV, Kang G, Kirkpatrick BD, Kirkwood CD, Mwenda JM, Parashar UD, Petri WA, Riddle MS, Steele AD, Thompson RL, Walson JL, Sanders JW, Mokdad AH, Murray CJL, Hay SI, Reiner RC. Rotavirus Vaccination and the Global Burden of Rotavirus Diarrhea Among Children Younger Than 5 Years. JAMA Pediatr 2018; 172:958-965. [PMID: 30105384 PMCID: PMC6233802 DOI: 10.1001/jamapediatrics.2018.1960] [Citation(s) in RCA: 554] [Impact Index Per Article: 79.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
IMPORTANCE Rotavirus infection is the global leading cause of diarrhea-associated morbidity and mortality among children younger than 5 years. OBJECTIVES To examine the extent of rotavirus infection among children younger than 5 years by country and the number of deaths averted because of the rotavirus vaccine. DESIGN, SETTING, AND PARTICIPANTS This report builds on findings from the Global Burden of Disease Study 2016, a cross-sectional study that measured diarrheal diseases and their etiologic agents. Models were used to estimate burden in data-sparse locations. EXPOSURE Diarrhea due to rotavirus infection. MAIN OUTCOMES AND MEASURES Rotavirus-associated mortality and morbidity by country and year and averted deaths attributable to the rotavirus vaccine by country. RESULTS Rotavirus infection was responsible for an estimated 128 500 deaths (95% uncertainty interval [UI], 104 500-155 600) among children younger than 5 years throughout the world in 2016, with 104 733 deaths occurring in sub-Saharan Africa (95% UI, 83 406-128 842). Rotavirus infection was responsible for more than 258 million episodes of diarrhea among children younger than 5 years in 2016 (95% UI, 193 million to 341 million), an incidence of 0.42 cases per child-year (95% UI, 0.30-0.53). Vaccine use is estimated to have averted more than 28 000 deaths (95% UI, 14 600-46 700) among children younger than 5 years, and expanded use of the rotavirus vaccine, particularly in sub-Saharan Africa, could have prevented approximately 20% of all deaths attributable to diarrhea among children younger than 5 years. CONCLUSIONS AND RELEVANCE Rotavirus-associated mortality has decreased markedly over time in part because of the introduction of the rotavirus vaccine. This study suggests that prioritizing vaccine introduction and interventions to reduce diarrhea-associated morbidity and mortality is necessary in the continued global reduction of rotavirus infection.
Collapse
Affiliation(s)
| | | | - Puja C. Rao
- Institute for Health Metrics and Evaluation, Seattle, Washington
| | - Shujin Cao
- Institute for Health Metrics and Evaluation, Seattle, Washington
| | | | - Tahmeed Ahmed
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - George Armah
- Noguchi Memorial Institute for Medical Research, Accra, Ghana
| | - Julie E. Bines
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia,Murdoch Children’s Research Institute, Department of Gastroenterology, Clinical Nutrition Royal Children's Hospital, Parkville, Melbourne, Victoria, Australia
| | | | | | - Gagandeep Kang
- Translational Health Science and Technology Institute, Faridabad, India
| | - Beth D. Kirkpatrick
- Department of Medicine, University of Vermont College of Medicine, Burlington
| | | | - Jason M. Mwenda
- World Health Organization Regional Office for Africa, Brazzaville, Republic of Congo
| | | | - William A. Petri
- Division of Infectious Diseases and International Health, Department of Internal Medicine, University of Virginia, Charlottesville
| | | | | | | | - Judd L. Walson
- Department of Global Health, University of Washington, Seattle,Department of Medicine, University of Washington, Seattle,Department of Pediatrics, University of Washington, Seattle,Department of Epidemiology, University of Washington, Seattle
| | - John W. Sanders
- Wake Forest University School of Medicine, Salem, North Carolina
| | - Ali H. Mokdad
- Institute for Health Metrics and Evaluation, Seattle, Washington
| | | | - Simon I. Hay
- Institute for Health Metrics and Evaluation, Seattle, Washington,Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Robert C. Reiner
- Institute for Health Metrics and Evaluation, Seattle, Washington
| |
Collapse
|
48
|
DeAntonio R, Amador S, Bunge EM, Eeuwijk J, Prado-Cohrs D, Nieto Guevara J, Rubio MDP, Ortega-Barria E. Vaccination herd effect experience in Latin America: a systematic literature review. Hum Vaccin Immunother 2018; 15:49-71. [PMID: 30230953 PMCID: PMC6363147 DOI: 10.1080/21645515.2018.1514225] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background: National pediatric vaccination programs have been introduced in Latin America (LatAm) to reduce the burden of diseases due to pathogens such as rotavirus, Haemophilus influenzae type b (Hib) and pneumococcus. Vaccination health benefits may extend to unvaccinated populations by reducing pathogen transmission. Understanding herd effect is important for implementation and assessment of vaccination programs. The objective was to conduct a systematic review of published epidemiological evidence of herd effect with Hib, rotavirus and pneumococcal conjugate vaccines (PCV) in LatAm. Methods: Searches were conducted in PubMed, Virtual Health Library (VHL), SciELO and SCOPUS databases, for studies reporting data on herd effect from Hib, rotavirus and PCV vaccination in LatAm, without age restriction. Searches were limited to articles published in English, Spanish or Portuguese (1990–2016). After screening and full-text review, articles meeting the selection criteria were included to be critically appraised following criteria for observational and interventional studies. The presence of a herd effect was defined as a significant decrease in incidence of disease, hospitalization, or mortality. Results: 3,465 unique articles were identified, and 23 were included (Hib vaccine n = 5, PCV n = 8, rotavirus vaccine n = 10). Most studies included children and/or adolescents (age range varied between studies). Studies in adults, including older adults (aged > 65 years), were limited. Few studies reported statistically significant reductions in disease incidence in age groups not targeted for vaccination. Hib-confirmed meningitis hospitalization decreased in children but herd effect could not be quantified. Some evidence of herd effect was identified for PCV and rotavirus vaccine in unvaccinated children. Evidence for herd effects due to PCV in adults was limited. Conclusion: After introduction of Hib, PCV and rotavirus vaccination in LatAm, reductions in morbidity/mortality have been reported in children not targeted for vaccination. However, due to methodological limitations (e.g. short post-vaccination periods and age range studied), there is currently insufficient evidence to quantify the herd effect in adult populations. More research and higher quality surveillance is needed to characterize herd effect of these vaccines in LatAm.
Collapse
Affiliation(s)
- Rodrigo DeAntonio
- a Centro de Vacunación Internacional S A CEVAXIN , Panama City , Panama
| | | | - Eveline M Bunge
- c Pallas Health Research and Consultancy BV , Rotterdam , the Netherlands
| | - Jennifer Eeuwijk
- c Pallas Health Research and Consultancy BV , Rotterdam , the Netherlands
| | | | | | | | | |
Collapse
|
49
|
Bar-Zeev N, King C, Phiri T, Beard J, Mvula H, Crampin AC, Heinsbroek E, Lewycka S, Tate JE, Parashar UD, Costello A, Mwansambo C, Heyderman RS, French N, Cunliffe NA. Impact of monovalent rotavirus vaccine on diarrhoea-associated post-neonatal infant mortality in rural communities in Malawi: a population-based birth cohort study. Lancet Glob Health 2018; 6:e1036-e1044. [PMID: 30103981 PMCID: PMC6088152 DOI: 10.1016/s2214-109x(18)30314-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 05/23/2018] [Accepted: 06/19/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND Rotavirus is a major contributor to child mortality. The effect of rotavirus vaccine on diarrhoea mortality has been estimated in middle-income but not low-income settings, where mortality is high and vaccine effectiveness in reducing admissions to hospital is lower. Empirical population-based mortality studies have not been done in any setting. Malawi introduced monovalent rotavirus vaccine (RV1) in October, 2012. We aimed to investigate the impact and effectiveness of the RV1 vaccine in reducing diarrhoea-associated mortality in infants aged 10-51 weeks. METHODS In this population-based cohort study, we included infants born between Jan 1, 2012, and June 1, 2015, in Mchinji, Central Malawi and analysed data on those surviving 10 weeks. Individual vaccination status was extracted from caregiver-held records or report at home visits at 4 months and 1 year of age. Survival to 1 year was confirmed at home visit, or cause of death ascertained by verbal autopsy. We assessed impact (1 minus mortality rate ratio following vs before vaccine introduction) using Poisson regression. Among vaccine-eligible infants (born from Sept 17, 2012), we assessed effectiveness (1 minus hazard ratio) using Cox regression. FINDINGS Between Jan 1, 2012, and June 1, 2015, we recruited 48 672 livebirths in Mchinji, among whom 38 518 were vaccine-eligible and 37 570 survived to age 10 weeks. Two-dose versus zero-dose effectiveness analysis included 28 141 infants, of whom 101 had diarrhoea-associated death before 1 year of age. Diarrhoea-associated mortality declined by 31% (95% CI 1-52; p=0·04) after RV1 introduction. Effectiveness against diarrhoea-mortality was 34% (95% CI -28 to 66; p=0·22). INTERPRETATION RV1 was associated with substantial reduction in diarrhoea-associated deaths among infants in this rural sub-Saharan African setting. These data add considerable weight to evidence showing the impact of rotavirus vaccine programmes. FUNDING Wellcome Trust and GlaxoSmithKline Biologicals.
Collapse
Affiliation(s)
- Naor Bar-Zeev
- Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK; Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Chichiri, Blantyre, Malawi; International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Tambosi Phiri
- Mai Mwana Project, Mchinji, Malawi; Parent and Child Health Initiative, Lilongwe, Malawi
| | - James Beard
- Mai Mwana Project, Mchinji, Malawi; London School of Hygiene & Tropical Medicine, London, UK
| | - Hazzie Mvula
- Malawi Epidemiology and Intervention Research Unit, formerly Karonga Prevention Study, Chilumba, Malawi
| | - Amelia C Crampin
- London School of Hygiene & Tropical Medicine, London, UK; Malawi Epidemiology and Intervention Research Unit, formerly Karonga Prevention Study, Chilumba, Malawi
| | - Ellen Heinsbroek
- Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK; Malawi Epidemiology and Intervention Research Unit, formerly Karonga Prevention Study, Chilumba, Malawi
| | - Sonia Lewycka
- Mai Mwana Project, Mchinji, Malawi; Institute for Global Health, University College London, London, UK
| | | | | | - Anthony Costello
- Child and Adolescent Health, World Health Organization, Geneva, Switzerland
| | - Charles Mwansambo
- Mai Mwana Project, Mchinji, Malawi; Parent and Child Health Initiative, Lilongwe, Malawi; Ministry of Health, Lilongwe, Malawi
| | - Robert S Heyderman
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Chichiri, Blantyre, Malawi; Division of Infection and Immunity, University College London, London, UK
| | - Neil French
- Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK; Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Chichiri, Blantyre, Malawi
| | - Nigel A Cunliffe
- Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK.
| |
Collapse
|
50
|
Cowley D, Nirwati H, Donato CM, Bogdanovic-Sakran N, Boniface K, Kirkwood CD, Bines JE. Molecular characterisation of rotavirus strains detected during a clinical trial of the human neonatal rotavirus vaccine (RV3-BB) in Indonesia. Vaccine 2018; 36:5872-5878. [PMID: 30145099 PMCID: PMC6143382 DOI: 10.1016/j.vaccine.2018.08.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 12/12/2022]
Abstract
Equine-like G3P[8] the major cause of gastroenteritis during RV3-BB efficacy trial. The Indonesian equine-like G3P[8] strain was genetically similar to Hungarian and Spanish strains. Equine-like G3P[8] strain is an emerging cause of gastroenteritis in Indonesia.
Background The RV3-BB human neonatal rotavirus vaccine aims to provide protection from severe rotavirus disease from birth. The aim of the current study was to characterise the rotavirus strains causing gastroenteritis during the Indonesian Phase IIb efficacy trial. Methods A randomized, double-blind placebo-controlled trial involving 1649 participants was conducted from January 2013 to July 2016 in Central Java and Yogyakarta, Indonesia. Participants received three doses of oral RV3-BB vaccine with the first dose given at 0–5 days after birth (neonatal schedule), or the first dose given at ∼8 weeks after birth (infant schedule), or placebo (placebo schedule). Stool samples from episodes of gastroenteritis were tested for rotavirus using EIA testing, positive samples were genotyped by RT-PCR. Full genome sequencing was performed on two representative rotavirus strains. Results There were 1110 episodes of acute gastroenteritis of any severity, 105 episodes were confirmed as rotavirus gastroenteritis by EIA testing. The most common genotype identified was G3P[8] (90/105), the majority (52/56) of severe (Vesikari score ≥11) rotavirus gastroenteritis episodes were due to the G3P[8] strain. Full genome analysis of two representative G3P[8] samples demonstrated the strain was an inter-genogroup reassortant, containing an equine-like G3 VP7, P[8] VP4 and a genogroup 2 backbone I2-R2-C2-M2-A2-N2-T2-E2-H2. The complete genome of the Indonesian equine-like G3P[8] strain demonstrated highest genetic identity to G3P[8] strains circulating in Hungary and Spain. Conclusions The dominant circulating strain during the Indonesian Phase IIb efficacy trial of the RV3-BB vaccine was an equine-like G3P[8] strain. The equine-like G3P[8] strain is an emerging cause of severe gastroenteritis in Indonesia and in other regions.
Collapse
Affiliation(s)
- Daniel Cowley
- Enteric Virus Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Rotavirus Program, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Hera Nirwati
- Department of Microbiology, Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Celeste M Donato
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Victoria, Australia
| | - Nada Bogdanovic-Sakran
- Enteric Virus Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Rotavirus Program, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Karen Boniface
- Enteric Virus Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Rotavirus Program, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Carl D Kirkwood
- Enteric Virus Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Julie E Bines
- Enteric Virus Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Rotavirus Program, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia; Department of Gastroenterology and Clinical Nutrition, Royal Children's Hospital, Parkville, Victoria, Australia.
| |
Collapse
|