1
|
Gong X, Huang Z, Zheng Y, Xiao W, Liu J, Lin S, Pan H, Chen J, Wu H, Wang W. Rotavirus vaccine effectiveness and coverage among children younger than 5 years old in Shanghai, China: A test-negative case control study. Vaccine 2025; 48:126731. [PMID: 39809089 DOI: 10.1016/j.vaccine.2025.126731] [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: 07/16/2024] [Revised: 12/29/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
OBJECTIVES The number of post-marketing studies assessing the vaccine effectiveness (VE) of the Lanzhou lamb rotavirus vaccine (LLR, licensed in 2000 exclusively in China) and the oral human attenuated pentavalent rotavirus vaccine (RotaTeq, licensed in China in 2018) in China is limited. METHODS A test-negative case-control study based on prospective surveillance was conducted among diarrhea patients aged 5 years and younger at five hospitals in Shanghai, China. Cases and controls were defined based on the results of real-time fluorescent quantitative reverse transcription polymerase chain reaction (rRT-PCR) of fecal samples for rotavirus. Both matched and unmatched case-control study designs were employed using logistic regression models, with adjustments for age at onset age and the rotavirus epidemic season. RESULTS In the LLR-specific analysis (247 cases, 2191 controls), the VE of partial LLR vaccination (2 doses) was 49.09 % (95 % CI: 1.69 % ∼ 73.64 %) in multivariate analyses. In the RotaTeq-specific analysis (42 cases and 523 controls), the VE of complete RotaTeq vaccination was 87.13 % (95 % CI: 45.87 % ∼ 96.94 %), 89.46 % (95 % CI: 55.03 % ∼ 97.53 %), and 85.69 % (95 % CI: 33.43 % ∼ 96.93 %) respectively in univariate, multivariate, and matched analyses, respectively. The vaccination coverage for any dose among 2893 patients with rotavirus-negative diarrhea born between 2011 and 2022 was 49.78 %. Following the licensure of RotaTeq in 2018, this coverage increased from 45.02 % to 61.77 %. CONCLUSIONS RotaTeq demonstrates a robust protective effectiveness, while LLR provides a certain level of protection against mild to moderate rotavirus diarrhea in children in Shanghai. For privately purchased (non-NIP) vaccines, we estimate that the coverage for rotavirus vaccines among children in Shanghai is high. Complete rotavirus vaccination is recommended for age-eligible children. Further post-marketing research on rotavirus vaccines is necessary to inform decision-making regarding the introduction of rotavirus vaccination in China.
Collapse
Affiliation(s)
- Xiaohuan Gong
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Division of Infectious Disease Control and Prevention, Shanghai Municipal Center of Disease Control and Prevention, Shanghai, China
| | - Zhuoying Huang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Division of Immunization Program, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Yaxu Zheng
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Division of Infectious Disease Control and Prevention, Shanghai Municipal Center of Disease Control and Prevention, Shanghai, China
| | - Wenjia Xiao
- Division of Infectious Disease Control and Prevention, Shanghai Municipal Center of Disease Control and Prevention, Shanghai, China
| | - Jiechen Liu
- Division of Immunization Program, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Sheng Lin
- Division of Infectious Disease Control and Prevention, Shanghai Municipal Center of Disease Control and Prevention, Shanghai, China
| | - Hao Pan
- Division of Infectious Disease Control and Prevention, Shanghai Municipal Center of Disease Control and Prevention, Shanghai, China
| | - Jian Chen
- Division of Infectious Disease Control and Prevention, Shanghai Municipal Center of Disease Control and Prevention, Shanghai, China
| | - Huanyu Wu
- Division of Infectious Disease Control and Prevention, Shanghai Municipal Center of Disease Control and Prevention, Shanghai, China
| | - Weibing Wang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Deniz Y, Demircioğlu B, Kuter-Emeklioğlu İ, Ekmen S, Sevınc E, Dogan E. Rotavirus Infections: A 2-Year Comprehensive Review in Admitted Pediastric Patients Amid Conflicting National Policies. Foodborne Pathog Dis 2024; 21:738-744. [PMID: 39185568 DOI: 10.1089/fpd.2024.0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024] Open
Abstract
Highlighting rotavirus (RV) as a significant food and waterborne pathogen, particularly affecting infants and children, causing serious gastrointestinal infections and dehydration, is important. It should be noted that there are significant debates regarding the effectiveness and outcomes of RV vaccination. In contrast to Turkey's nonmandatory vaccination policy, many developed countries implement mandatory vaccination policies, raising questions about their impact on disease prevalence and healthcare expenditures. Our study aims to comprehensively understand RV infections in Turkey and compare them with countries that have mandatory vaccination policies. We found similar, and even better, hospitalization rates, length of hospital stays, and laboratory parameters demonstrating the effectiveness of Turkey's independent vaccination approach. These findings contribute valuable insights to global vaccination strategies and disease control.
Collapse
Affiliation(s)
- Yusuf Deniz
- Department of Pediatrics, Faculty of Medicine Training and Research Hospital, Karabuk University, Karabuk, Turkey
| | - Burak Demircioğlu
- Department of Pediatrics, Faculty of Medicine Training and Research Hospital, Karabuk University, Karabuk, Turkey
| | - İrem Kuter-Emeklioğlu
- Department of Pediatrics, Faculty of Medicine Training and Research Hospital, Karabuk University, Karabuk, Turkey
| | - Sadrettin Ekmen
- Department of Pediatrics, Faculty of Medicine Training and Research Hospital, Neonatal Intensive Care Unit, Karabuk University, Karabuk, Turkey
| | - Eylem Sevınc
- Department of Pediatrics, Faculty of Medicine Training and Research Hospital, Pediatric Gastroenterology Hepatology & Nutrition, Karabuk University, Karabuk, Turkey
| | - Erkan Dogan
- Department of Pediatrics, Faculty of Medicine Training and Research Hospital, Karabuk University, Karabuk, Turkey
| |
Collapse
|
3
|
Peña-Gil N, Randazzo W, Carmona-Vicente N, Santiso-Bellón C, Cárcamo-Cálvo R, Navarro-Lleó N, Monedero V, Yebra MJ, Buesa J, Gozalbo-Rovira R, Rodríguez-Díaz J. Culture of Human Rotaviruses in Relevant Models Shows Differences in Culture-Adapted and Nonculture-Adapted Strains. Int J Mol Sci 2023; 24:17362. [PMID: 38139191 PMCID: PMC10743750 DOI: 10.3390/ijms242417362] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Rotavirus (RV) is the leading cause of acute gastroenteritis (AGE) in children under 5 years old worldwide, and several studies have demonstrated that histo-blood group antigens (HBGAs) play a role in its infection process. In the present study, human stool filtrates from patients diagnosed with RV diarrhea (genotyped as P[8]) were used to infect differentiated Caco-2 cells (dCaco-2) to determine whether such viral strains of clinical origin had the ability to replicate in cell cultures displaying HBGAs. The cell culture-adapted human RV Wa model strain (P[8] genotype) was used as a control. A time-course analysis of infection was conducted in dCaco-2 at 1, 24, 48, 72, and 96 h. The replication of two selected clinical isolates and Wa was further assayed in MA104, undifferentiated Caco-2 (uCaco-2), HT29, and HT29-M6 cells, as well as in monolayers of differentiated human intestinal enteroids (HIEs). The results showed that the culture-adapted Wa strain replicated more efficiently in MA104 cells than other utilized cell types. In contrast, clinical virus isolates replicated more efficiently in dCaco-2 cells and HIEs. Furthermore, through surface plasmon resonance analysis of the interaction between the RV spike protein (VP8*) and its glycan receptor (the H antigen), the V7 RV clinical isolate showed 45 times better affinity compared to VP8* from the Wa strain. These findings support the hypothesis that the differences in virus tropism between clinical virus isolates and RV Wa could be a consequence of the different HBGA contents on the surface of the cell lines employed. dCaco-2, HT29, and HT29M6 cells and HIEs display HBGAs on their surfaces, whereas MA104 and uCaco-2 cells do not. These results indicate the relevance of using non-cell culture-adapted human RV to investigate the replication of rotavirus in relevant infection models.
Collapse
Affiliation(s)
- Nazaret Peña-Gil
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (N.P.-G.); (N.C.-V.); (C.S.-B.); (R.C.-C.); (N.N.-L.); (J.B.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Walter Randazzo
- Department of Preservation and Food Safety Technologies, IATA-CSIC, Av. Agustín Escardino 7, 46980 Paterna, Spain;
| | - Noelia Carmona-Vicente
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (N.P.-G.); (N.C.-V.); (C.S.-B.); (R.C.-C.); (N.N.-L.); (J.B.)
| | - Cristina Santiso-Bellón
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (N.P.-G.); (N.C.-V.); (C.S.-B.); (R.C.-C.); (N.N.-L.); (J.B.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Roberto Cárcamo-Cálvo
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (N.P.-G.); (N.C.-V.); (C.S.-B.); (R.C.-C.); (N.N.-L.); (J.B.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Noemi Navarro-Lleó
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (N.P.-G.); (N.C.-V.); (C.S.-B.); (R.C.-C.); (N.N.-L.); (J.B.)
| | - Vicente Monedero
- Department of Biotechnology, IATA-CSIC, Av. Agustín Escardino 7, 46980 Paterna, Spain; (V.M.); (M.J.Y.)
| | - María J. Yebra
- Department of Biotechnology, IATA-CSIC, Av. Agustín Escardino 7, 46980 Paterna, Spain; (V.M.); (M.J.Y.)
| | - Javier Buesa
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (N.P.-G.); (N.C.-V.); (C.S.-B.); (R.C.-C.); (N.N.-L.); (J.B.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Roberto Gozalbo-Rovira
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (N.P.-G.); (N.C.-V.); (C.S.-B.); (R.C.-C.); (N.N.-L.); (J.B.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Jesús Rodríguez-Díaz
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; (N.P.-G.); (N.C.-V.); (C.S.-B.); (R.C.-C.); (N.N.-L.); (J.B.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| |
Collapse
|
4
|
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]
|
5
|
Du Y, Chen C, Zhang X, Yan D, Jiang D, Liu X, Yang M, Ding C, Lan L, Hecht R, Zhu C, Yang S. Global burden and trends of rotavirus infection-associated deaths from 1990 to 2019: an observational trend study. Virol J 2022; 19:166. [PMID: 36266651 PMCID: PMC9585833 DOI: 10.1186/s12985-022-01898-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 10/03/2022] [Indexed: 11/12/2022] Open
Abstract
Background Rotavirus is the leading global pathogen of diarrhea-associated mortality and poses a great threat to public health in all age groups. This study aimed to explore the global burden and 30-year change patterns of rotavirus infection-associated deaths. Methods Based on the Global Burden of Disease 2019 Study (GBD 2019), we analyzed the age-standardized death rate (ASDR) of rotavirus infection by sex, geographical region, and sociodemographic index (SDI) from 1990 to 2019. A Joinpoint regression model was used to analyze the global trends in rotavirus infection over the 30 years, SaTScan software was used to detect the spatial and temporal aggregations, and a generalized linear model to explore the relationship between sociodemographic factors and death rates of rotavirus infection. Results Globally, rotavirus infection was the leading cause of diarrheal deaths, accounting for 19.11% of deaths from diarrhea in 2019. Rotavirus caused a higher death burden in African, Oceanian, and South Asian countries in the past three decades. The ASDR of rotavirus declined from 11.39 (95% uncertainty interval [95% UI] 5.46–19.48) per 100,000 people in 1990 to 3.41 (95% UI 1.60–6.01) per 100,000 people in 2019, with an average annual percentage change (AAPC) (− 4.07%, P < 0.05). However, a significant uptrend was found in high-income North America (AAPC = 1.79%, P < 0.05). The death rate was the highest among children under 5 years worldwide. However, the death rates of elderly individuals over 70 years were higher than those of children under 5 years in 2019 among high, high-middle, middle, and low-middle SDI regions. Current health expenditure, gross domestic product per capita, and the number of physicians per 1000 people were significantly negatively correlated with death rates of rotavirus. Conclusions Although the global trends in the rotavirus burden have decreased substantially over the past three decades, the burden of rotavirus remained high in Africa, Oceania, and South Asia. Children under 5 years and elderly individuals over 70 years were the populations most at risk for rotavirus infection-associated deaths, especially elderly individuals over 70 years in relatively high SDI regions. More attention should be paid to these areas and populations, and effective public health policies should be implemented in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s12985-022-01898-9.
Collapse
Affiliation(s)
- Yuxia Du
- Department of Public Health, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, China
| | - Can Chen
- Department of Public Health, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, China
| | - Xiaobao Zhang
- Department of Public Health, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, China
| | - Danying Yan
- Department of Public Health, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, China
| | - Daixi Jiang
- Department of Public Health, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, China
| | - Xiaoxiao Liu
- Department of Public Health, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, China
| | - Mengya Yang
- Department of Public Health, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, China
| | - Cheng Ding
- Department of Public Health, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, China
| | - Lei Lan
- Department of Public Health, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, China
| | - Robert Hecht
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, 06520, USA
| | - Changtai Zhu
- Department of Laboratory Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Shigui Yang
- Department of Public Health, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, China.
| |
Collapse
|
6
|
Mijatovic-Rustempasic S, Jaimes J, Perkins C, Ward ML, Esona MD, Gautam R, Lewis J, Sturgeon M, Panjwani J, Bloom GA, Miller S, Reisdorf E, Riley AM, Pence MA, Dunn J, Selvarangan R, Jerris RC, DeGroat D, Parashar UD, Cortese MM, Bowen MD. Rotavirus Strain Trends in United States, 2009-2016: Results from the National Rotavirus Strain Surveillance System (NRSSS). Viruses 2022; 14:1775. [PMID: 36016397 PMCID: PMC9414880 DOI: 10.3390/v14081775] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Before the introduction of vaccines, group A rotaviruses (RVA) were the leading cause of acute gastroenteritis in children worldwide. The National Rotavirus Strain Surveillance System (NRSSS) was established in 1996 by the Centers for Disease Control and Prevention (CDC) to perform passive RVA surveillance in the USA. We report the distribution of RVA genotypes collected through NRSSS during the 2009-2016 RVA seasons and retrospectively examine the genotypes detected through the NRSSS since 1996. During the 2009-2016 RVA seasons, 2134 RVA-positive fecal specimens were sent to the CDC for analysis of the VP7 and VP4 genes by RT-PCR genotyping assays and sequencing. During 2009-2011, RVA genotype G3P[8] dominated, while G12P[8] was the dominant genotype during 2012-2016. Vaccine strains were detected in 1.7% of specimens and uncommon/unusual strains, including equine-like G3P[8] strains, were found in 1.9%. Phylogenetic analyses showed limited VP7 and VP4 sequence variation within the common genotypes with 1-3 alleles/lineages identified per genotype. A review of 20 years of NRSSS surveillance showed two changes in genotype dominance, from G1P[8] to G3P[8] and then G3P[8] to G12P[8]. A better understanding of the long-term effects of vaccine use on epidemiological and evolutionary dynamics of circulating RVA strains requires continued surveillance.
Collapse
Affiliation(s)
- Slavica Mijatovic-Rustempasic
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mail Stop G-04, Atlanta, GA 30329, USA
| | - Jose Jaimes
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mail Stop G-04, Atlanta, GA 30329, USA
| | - Charity Perkins
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mail Stop G-04, Atlanta, GA 30329, USA
| | - M. Leanne Ward
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mail Stop G-04, Atlanta, GA 30329, USA
| | - Mathew D. Esona
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mail Stop G-04, Atlanta, GA 30329, USA
| | - Rashi Gautam
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mail Stop G-04, Atlanta, GA 30329, USA
| | - Jamie Lewis
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mail Stop G-04, Atlanta, GA 30329, USA
| | - Michele Sturgeon
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mail Stop G-04, Atlanta, GA 30329, USA
| | - Junaid Panjwani
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mail Stop G-04, Atlanta, GA 30329, USA
| | - Gail A. Bloom
- Indiana University Health Pathology Laboratory, Indiana University, 350 West 11th Street, Indianapolis, IN 46202, USA
| | - Steve Miller
- UCSF Clinical Microbiology Laboratory, 185 Berry St, Suite 290, San Francisco, CA 94107, USA
| | - Erik Reisdorf
- Wisconsin State Laboratory of Hygiene, 2601 Agriculture Drive, Madison, WI 53718, USA
| | - Ann Marie Riley
- Infectious Disease Diagnostic Laboratory, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
| | - Morgan A. Pence
- Cook Children’s Medical Center, 801 Seventh Ave., Fort Worth, TX 76104, USA
| | - James Dunn
- Medical Microbiology and Virology, Department of Pathology, Texas Children’s Hospital, 6621 Fannin Street, Suite AB1195, Houston, TX 77030, USA
| | | | - Robert C. Jerris
- Children’s Healthcare of Atlanta, 1405 Clifton Rd, Atlanta, GA 30329, USA
| | - Dona DeGroat
- Seattle Children’s Hospital, 5801 Sand Point Way NE, Seattle, WA 98105, USA
| | - Umesh D. Parashar
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mail Stop G-04, Atlanta, GA 30329, USA
| | - Margaret M. Cortese
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mail Stop G-04, Atlanta, GA 30329, USA
| | - Michael D. Bowen
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mail Stop G-04, Atlanta, GA 30329, USA
| |
Collapse
|
7
|
Kumar M, James MM, Kumawat M, Nabi B, Sharma P, Pal N, Shubham S, Tiwari RR, Sarma DK, Nagpal R. Aging and Microbiome in the Modulation of Vaccine Efficacy. Biomedicines 2022; 10:biomedicines10071545. [PMID: 35884849 PMCID: PMC9313064 DOI: 10.3390/biomedicines10071545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 12/29/2022] Open
Abstract
From infancy through to old age, the microbiome plays an important role in modulating the host-immune system. As we age, our immune system and our gut microbiota change significantly in composition and function, which is linked to an increased vulnerability to infectious diseases and a decrease in vaccine responses. Our microbiome remains largely stable throughout adulthood; however, aging causes a major shift in the composition and function of the gut microbiome, as well as a decrease in diversity. Considering the critical role of the gut microbiome in the host-immune system, it is important to address, prevent, and ameliorate age-related dysbiosis, which could be an effective strategy for preventing/restoring functional deficits in immune responses as we grow older. Several factors, such as the host’s genetics and nutritional state, along with the gut microbiome, can influence vaccine efficacy or reaction. Emerging evidence suggests that the microbiome could be a significant determinant of vaccine immunity. Physiological mechanisms such as senescence, or the steady loss of cellular functions, which affect the aging process and vaccination responses, have yet to be comprehended. Recent studies on several COVID-19 vaccines worldwide have provided a considerable amount of data to support the hypothesis that aging plays a crucial role in modulating COVID-19 vaccination efficacy across different populations.
Collapse
Affiliation(s)
- Manoj Kumar
- National Institute for Research in Environmental Health, Bhopal 462030, India; (M.K.); (M.M.J.); (M.K.); (P.S.); (N.P.); (S.S.); (R.R.T.)
| | - Meenu Mariya James
- National Institute for Research in Environmental Health, Bhopal 462030, India; (M.K.); (M.M.J.); (M.K.); (P.S.); (N.P.); (S.S.); (R.R.T.)
| | - Manoj Kumawat
- National Institute for Research in Environmental Health, Bhopal 462030, India; (M.K.); (M.M.J.); (M.K.); (P.S.); (N.P.); (S.S.); (R.R.T.)
| | - Bilkees Nabi
- Department of Biochemistry and Biochemical Engineering, Sam Higginbottom University of Agriculture, Technology and Sciences, Allahabad 211007, India;
| | - Poonam Sharma
- National Institute for Research in Environmental Health, Bhopal 462030, India; (M.K.); (M.M.J.); (M.K.); (P.S.); (N.P.); (S.S.); (R.R.T.)
| | - Namrata Pal
- National Institute for Research in Environmental Health, Bhopal 462030, India; (M.K.); (M.M.J.); (M.K.); (P.S.); (N.P.); (S.S.); (R.R.T.)
| | - Swasti Shubham
- National Institute for Research in Environmental Health, Bhopal 462030, India; (M.K.); (M.M.J.); (M.K.); (P.S.); (N.P.); (S.S.); (R.R.T.)
| | - Rajnarayan R. Tiwari
- National Institute for Research in Environmental Health, Bhopal 462030, India; (M.K.); (M.M.J.); (M.K.); (P.S.); (N.P.); (S.S.); (R.R.T.)
| | - Devojit Kumar Sarma
- National Institute for Research in Environmental Health, Bhopal 462030, India; (M.K.); (M.M.J.); (M.K.); (P.S.); (N.P.); (S.S.); (R.R.T.)
- Correspondence: (D.K.S.); (R.N.)
| | - Ravinder Nagpal
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL 32302, USA
- Correspondence: (D.K.S.); (R.N.)
| |
Collapse
|
8
|
Meki CD, Ncube EJ, Voyi K. Community-level interventions for mitigating the risk of waterborne diarrheal diseases: a systematic review. Syst Rev 2022; 11:73. [PMID: 35436979 PMCID: PMC9016942 DOI: 10.1186/s13643-022-01947-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 04/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Waterborne diarrhea diseases are among the leading causes of morbidity and mortality globally. These diseases can be mitigated by implementing various interventions. We reviewed the literature to identify available interventions to mitigate the risk of waterborne diarrheal diseases. METHODS We conducted a systematic database review of CINAHL (Cumulative Index to Nursing and Allied Health Literature), PubMed, Web of Science Core Collection, Cochrane library, Scopus, African Index Medicus (AIM), and LILACS (Latin American and Caribbean Health Sciences Literature). Our search was limited to articles published between 2009 and 2020. We conducted the review using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement checklist. The identified studies were qualitatively synthesized. RESULTS Our initial search returned 28 773 articles of which 56 studies met the inclusion criteria. The included studies reported interventions, including vaccines for rotavirus disease (monovalent, pentavalent, and Lanzhou lamb vaccine); enhanced water filtration for preventing cryptosporidiosis, Vi polysaccharide for typhoid; cholera 2-dose vaccines, water supply, water treatment and safe storage, household disinfection, and hygiene promotion for controlling cholera outbreaks. CONCLUSION We retrieved few studies on interventions against waterborne diarrheal diseases in low-income countries. Interventions must be specific to each type of waterborne diarrheal disease to be effective. Stakeholders must ensure collaboration in providing and implementing multiple interventions for the best outcomes. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42020190411 .
Collapse
Affiliation(s)
- Chisala D. Meki
- University of Zambia, School of Public Health, University of Zambia, P O. BOX 50110, Lusaka, Zambia
- School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
| | - Esper J. Ncube
- School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
- Rand Water, Johannesburg, South Africa
| | - Kuku Voyi
- School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
9
|
Abstract
BACKGROUND Rotavirus causes 215,000 deaths from severe childhood diarrhea annually. Concerns exist that a monovalent vaccine (RV1) and a pentavalent vaccine (RV5) may be less effective against rotavirus strains not contained in the vaccines. We estimated the vaccine effectiveness (VE) of RV1 and RV5 against severe rotavirus gastroenteritis caused by vaccine (homotypic) and nonvaccine (partially and fully heterotypic) strains. METHODS After conducting a systematic review, we meta-analyzed 31 case-control studies (N = 27,293) conducted between 2006 and 2020 using a random-effects regression model. RESULTS In high-income countries, RV1 VE was 10% lower against partially heterotypic (P = 0.04) and fully heterotypic (P = 0.10) compared with homotypic strains (homotypic VE: 90% [95% confidence intervals (CI): 82-94]; partially heterotypic VE: 79% [95% CI: 71-85]; fully heterotypic VE: 80% [95% CI: 65-88]). In middle-income countries, RV1 VE was 14-16% lower against partially heterotypic (P = 0.06) and fully heterotypic (P = 0.04) compared with homotypic strains (homotypic VE: 81% [95% CI: 69-88]; partially heterotypic VE: 67% [95% CI: 54-76]; fully heterotypic VE: 65% [95% CI: 51-75]). Strain-specific RV5 VE differences were less pronounced, and primarily derived from high-income countries. Limited data were available from low-income countries. CONCLUSIONS Vaccine effectiveness of RV1 and RV5 was somewhat lower against nonvaccine than vaccine strains. Ongoing surveillance is important to continue long-term monitoring for strain replacement, particularly in low-income settings where data are limited.
Collapse
|
10
|
Chow EJ, Tenforde MW, Rolfes MA, Lee B, Chodisetty S, Ramirez JA, Fry AM, Patel MM. Differentiating severe and non-severe lower respiratory tract illness in patients hospitalized with influenza: Development of the Influenza Disease Evaluation and Assessment of Severity (IDEAS) scale. PLoS One 2021; 16:e0258482. [PMID: 34673782 PMCID: PMC8530291 DOI: 10.1371/journal.pone.0258482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/28/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Experimental studies have shown that vaccination can reduce viral replication to attenuate progression of influenza-associated lower respiratory tract illness (LRTI). However, clinical studies are conflicting, possibly due to use of non-specific outcomes reflecting a mix of large and small airway LRTI lacking specificity for acute lung or organ injury. METHODS We developed a global ordinal scale to differentiate large and small airway LRTI in hospitalized adults with influenza using physiologic features and interventions (PFIs): vital signs, laboratory and radiographic findings, and clinical interventions. We reviewed the literature to identify common PFIs across 9 existing scales of pneumonia and sepsis severity. To characterize patients using this scale, we applied the scale to an antiviral clinical trial dataset where these PFIs were measured through routine clinical care in adults hospitalized with influenza-associated LRTI during the 2010-2013 seasons. RESULTS We evaluated 12 clinical parameters among 1020 adults; 210 (21%) had laboratory-confirmed influenza, with a median severity score of 4.5 (interquartile range, 2-8). Among influenza cases, median age was 63 years, 20% were hospitalized in the prior 90 days, 50% had chronic obstructive pulmonary disease, and 22% had congestive heart failure. Primary influencers of higher score included pulmonary infiltrates on imaging (48.1%), heart rate ≥110 beats/minute (41.4%), oxygen saturation <93% (47.6%) and respiratory rate >24 breaths/minute (21.0%). Key PFIs distinguishing patients with severity < or ≥8 (upper quartile) included infiltrates (27.1% vs 90.0%), temperature ≥ 39.1°C or <36.0°C (7.1% vs 27.1%), respiratory rate >24 breaths/minute (7.9% vs 47.1%), heart rate ≥110 beats/minute (29.3% vs 65.7%), oxygen saturation <90% (14.3% vs 31.4%), white blood cell count >15,000 (5.0% vs 27.2%), and need for invasive or non-invasive mechanical ventilation (2.1% vs 15.7%). CONCLUSION We developed a scale in adults hospitalized with influenza-associated LRTI demonstrating a broad distribution of physiologic severity which may be useful for future studies evaluating the disease attenuating effects of influenza vaccination or other therapeutics.
Collapse
Affiliation(s)
- Eric J. Chow
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- Epidemic Intelligence Service, Center for Surveillance, Epidemiology and Laboratory Services, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Mark W. Tenforde
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- Epidemic Intelligence Service, Center for Surveillance, Epidemiology and Laboratory Services, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Melissa A. Rolfes
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Benjamin Lee
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Shreya Chodisetty
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Julio A. Ramirez
- Division of Infectious Diseases, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Alicia M. Fry
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Manish M. Patel
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| |
Collapse
|
11
|
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
|
12
|
Sun ZW, Fu Y, Lu HL, Yang RX, Goyal H, Jiang Y, Xu HG. Association of Rotavirus Vaccines With Reduction in Rotavirus Gastroenteritis in Children Younger Than 5 Years: A Systematic Review and Meta-analysis of Randomized Clinical Trials and Observational Studies. JAMA Pediatr 2021; 175:e210347. [PMID: 33970192 PMCID: PMC8111566 DOI: 10.1001/jamapediatrics.2021.0347] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
IMPORTANCE Rotavirus vaccines have been introduced worldwide, and the clinical association of different rotavirus vaccines with reduction in rotavirus gastroenteritis (RVGE) after introduction are noteworthy. OBJECTIVE To evaluate the comparative benefit, risk, and immunogenicity of different rotavirus vaccines by synthesizing randomized clinical trials (RCTs) and observational studies. DATA SOURCES Relevant studies published in 4 databases: Embase, PubMed, the Cochrane Library, and Web of Science were searched until July 1, 2020, using search terms including "rotavirus" and "vaccin*." STUDY SELECTION Randomized clinical trials and cohort and case-control studies involving more than 100 children younger than 5 years that reported the effectiveness, safety, or immunogenicity of rotavirus vaccines were included. DATA EXTRACTION AND SYNTHESIS A random-effects model was used to calculate relative risks (RRs), odds ratios (ORs), risk differences, and 95% CIs. Adjusted indirect treatment comparison was performed to assess the differences in the protection of Rotarix and RotaTeq. MAIN OUTCOMES AND MEASURES The primary outcomes were RVGE, severe RVGE, and RVGE hospitalization. Safety-associated outcomes involved serious adverse events, intussusception, and mortality. RESULTS A meta-analysis of 20 RCTs and 38 case-control studies revealed that Rotarix (RV1) significantly reduced RVGE (RR, 0.316 [95% CI, 0.224-0.345]) and RVGE hospitalization risk (OR, 0.347 [95% CI, 0.279-0.432]) among children fully vaccinated; RotaTeq (RV5) had similar outcomes (RVGE: RR, 0.350 [95% CI, 0.275-0.445]; RVGE hospitalization risk: OR, 0.272 [95% CI, 0.197-0.376]). Rotavirus vaccines also demonstrated higher protection against severe RVGE. Additionally, no significant differences in the protection of RV1 and RV5 against rotavirus disease were noted in adjusted indirect comparisons. Moderate associations were found between reduced RVGE risk and Rotavac (RR, 0.664 [95% CI, 0.548-0.804]), Rotasiil (RR, 0.705 [95% CI, 0.605-0.821]), and Lanzhou lamb rotavirus vaccine (RR, 0.407 [95% CI, 0.332-0.499]). All rotavirus vaccines demonstrated no risk of serious adverse events. A positive correlation was also found between immunogenicity and vaccine protection (eg, association of RVGE with RV1: coefficient, -1.599; adjusted R2, 99.7%). CONCLUSIONS AND RELEVANCE The high protection and low risk of serious adverse events for rotavirus vaccines in children who were fully vaccinated emphasized the importance of worldwide introduction of rotavirus vaccination. Similar protection provided by Rotarix and RotaTeq relieves the pressure of vaccines selection for health care authorities.
Collapse
Affiliation(s)
- Zi-Wei Sun
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Fu
- Department of Pathology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hai-Ling Lu
- Department of Laboratory Medicine, Yancheng Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, China
| | - Rui-Xia Yang
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hemant Goyal
- The Wright Center of Graduate Medical Education, Scranton, Pennsylvania
| | - Ye Jiang
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hua-Guo Xu
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
13
|
Lu MC, Shia BC, Kao YW, Lin SC, Wang CY, Lin WC, Chen SY. The impact of rotavirus vaccination in the prevalence of gastroenteritis and comorbidities among children after suboptimal rotavirus vaccines implementation in Taiwan: A population-based study. Medicine (Baltimore) 2021; 100:e25925. [PMID: 34160381 PMCID: PMC8238267 DOI: 10.1097/md.0000000000025925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 04/19/2021] [Indexed: 01/04/2023] Open
Abstract
In Taiwan, rotavirus vaccination was implemented in 2006 in the private sector. The population-based impact of rotavirus vaccination on gastroenteritis and comorbidities of children remains under-investigated.We analyzed the annual prevalence of rotavirus-related disease, including gastroenteritis, convulsions, epilepsy, type I diabetes mellitus, intussusception, and biliary atresia among children under 5 years of age. Data were collected from Taiwan's National Health Insurance Research Database, a nationwide population-based database. A 16-year retrospective cohort study was conducted between 2000 and 2015.Among children <5 years of age, the prevalence of gastroenteritis decreased after 2012 (44,259.69 per 100 thousands) and remained lower through 2015 (39,931.11per 100 thousands, P < .001). The prevalence of convulsions rose steadily and significantly from 2007 (775.90 per 100 thousands) to 2015 (962.17 per 100 thousands, P < .001). The prevalence of epilepsy decreased significantly until reaching a nadir in 2013 (from 501.56 to 293.53 per 100 thousands, P < .001). The prevalence of biliary atresia tended upward, and surged suddenly in 2007 with a peak in 2013 (18.74 per 100 thousands). Among infants (<1 year of age) from 2000 to 2015, the prevalence of gastroenteritis declined steadily, and more rapidly after 2007 (22,513 to 17,285 per 100 thousands).In Taiwan, after introducing rotavirus vaccination, gastroenteritis in young children decreased, especially in infancy. However, gastroenteritis is still common in children, given other emerging pathogens. Our results highlight the impact of rotavirus vaccines on children's health in Taiwan and provide indications for future preventive medicine and healthcare strategies in children.
Collapse
Affiliation(s)
- Meng-Che Lu
- Division of Allergy, Asthma and Immunology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City
| | - Ben-Chang Shia
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, New Taipei City
| | - Yi-Wei Kao
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, New Taipei City
- Research Center of Big Data, College of management
| | - Sheng-Chieh Lin
- Division of Allergy, Asthma and Immunology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei
| | - Chuan-Yu Wang
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei
- Division of Pediatric Neurology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City
| | - Wen-Chuan Lin
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei
- Division of Pediatric Infectious Disease, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City
| | - Shih-Yen Chen
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| |
Collapse
|
14
|
Abstract
Vaccines are the most effective means available for preventing infectious diseases. However, vaccine-induced immune responses are highly variable between individuals and between populations in different regions of the world. Understanding the basis of this variation is, thus, of fundamental importance to human health. Although the factors that are associated with intra- and inter-population variation in vaccine responses are manifold, emerging evidence points to a key role for the gut microbiome in controlling immune responses to vaccination. Much of this evidence comes from studies in mice, and causal evidence for the impact of the microbiome on human immunity is sparse. However, recent studies on vaccination in subjects treated with broad-spectrum antibiotics have provided causal evidence and mechanistic insights into how the microbiota controls immune responses in humans.
Collapse
|
15
|
Wang Y, Li J, Dai P, Liu P, Zhu F. Effectiveness of the oral human attenuated pentavalent rotavirus vaccine (RotaTeq™) postlicensure: a meta-analysis-2006-2020. Expert Rev Vaccines 2021; 20:437-448. [PMID: 33709863 DOI: 10.1080/14760584.2021.1902808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Rotavirus (RV), which causes RV-associated gastroenteritis (RVGE), has accounted for considerable morbidity. We aimed to assess the effectiveness (VE) of the oral pentavalent RV vaccine (RotaTeq™) in real-world settings in children and infants with gastroenteritis. METHODS We performed a systematic search for peer-reviewed studies published between 1 January 2006 and 1 May 2020 and a meta-analysis to calculate the VE of RotaTeq™ vaccine. The primary outcome was the pooled three-dose vaccine VE. Stratified analysis of the vaccine VEs was performed according to dosages, study design, population age, socioeconomic status (SES), introduction condition, control group types, outcomes of RV disease, and RV strains. RESULTS After screening 2359 unique records, 28 studies were included and meta-analyzed. The overall VE estimate was 84% (95% confidence interval [CI], 80-87%). Stratified analyses revealed a nonnegligible impact of factors such as study design and SES. Other factors did not show great impart to VE with no significant differences between groups. CONCLUSIONS RotaTeq™ is effective against RV infection, especially in high-income countries. Adopting suitable study methods and expansion of RV surveillance in low-income regions is crucial to assess VE in real-life settings and provide feasible vaccine regimens to improve vaccine VE.
Collapse
Affiliation(s)
- Yuxiao Wang
- School of Public Health, Southeast University, Nanjing, China
| | - Jingxin Li
- Vaccine Clinical Evaluation Department, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Pinyuan Dai
- School of Public Health, Southeast University, Nanjing, China
| | - Pei Liu
- School of Public Health, Southeast University, Nanjing, China
| | - Fengcai Zhu
- Vaccine Clinical Evaluation Department, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| |
Collapse
|
16
|
The impact of shared decision making with patient decision aids on the rotavirus vaccination rate in children: A randomized controlled trial. Prev Med 2020; 141:106244. [PMID: 32891678 DOI: 10.1016/j.ypmed.2020.106244] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 08/07/2020] [Accepted: 08/29/2020] [Indexed: 12/14/2022]
Abstract
Rotavirus vaccination reduces the incidence and severity of acute gastroenteritis due to rotavirus infection. However, because of a lack of understanding and private payment for the rotavirus vaccine, the rotavirus vaccination rate is still low in some countries. We intended to assess the impact of shared decision-making (SDM) with the assistance of patient decision aids (PDAs) on the rotavirus vaccination rate, and the knowledge, confidence, and congruence of value among baby's parents when decision-making. The study was a two-group, outcome assessor-blind, randomized, controlled trial. The families of 1-month-old infants for routine vaccination were enrolled; they were divided randomly into non-SDM and SDM groups. The influence of SDM on the acceptance of rotavirus vaccination was assessed when their infants were 2 months old. Outcome measures were decisional conflict, decision-making difficulties, and rotavirus vaccine knowledge, and the overall rotavirus vaccination rate. The study enrolled 180 participants. SDM, parents' education level, and rotavirus vaccination of a previous child were variables that influenced acceptance of rotavirus vaccination. The SDM group scored significantly higher for understanding the information on the oral rotavirus vaccine than the non-SDM group, which helped them to decide whether to vaccinate the baby against rotavirus. The rotavirus vaccination rate was 16.7% higher in the SDM group than the non-SDM group. SDM assisted with PDAs gives more information and helps infants' families understand what they need, reduces their decision conflict, and increases their baby's vaccination against rotavirus, which promotes public health. The clinical trial is registered at ClinicalTrials.gov (NCT03804489).
Collapse
|
17
|
Hajela N, Chattopadhyay S, Nair GB, Ganguly NK. Intestinal microbiota and vaccine efficacy in children from resource poor settings - potential impact for the usefulness of probiotics? Benef Microbes 2020; 11:319-328. [PMID: 32720834 DOI: 10.3920/bm2019.0155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Developing countries continue to contribute significantly to the global burden of childhood mortality due to infectious diseases. Infections leading to diseases like diarrhoea, pneumonia and meningitis account for millions of deaths annually. Most of these diseases are preventable by vaccination and therefore global vaccination rates have risen substantially with clear benefits. But paradoxically, the vaccines have demonstrated lower immunogenicity in developing countries as compared to their industrialised counterparts. Malnutrition in resource poor settings along with repeated polymicrobial infections at early age are some of the reasons for the differences in vaccine efficacy in different settings. Recent studies indicate that the gastrointestinal microbiota possibly influences maturation of immune system as well as vaccine efficacy. In this review we discuss evidences from in vitro, animal and human studies showing that probiotics can positively modulate gut microbiota composition and exert immunomodulatory effects on the host. We also discuss how they should be evaluated for their ability to improve vaccine performance especially in low resource settings.
Collapse
Affiliation(s)
- N Hajela
- Gut Microbiota and Probiotic Science Foundation (India), M-4, Level one, South Extension Part - Ii, New Delhi 110049, India
| | - S Chattopadhyay
- Rajiv Gandhi Centre for Biotechnology, Thycaud Post, Poojappura, Thiruvananthapuram, Trivandrum, 695014 Kerala, India
| | - G B Nair
- Rajiv Gandhi Centre for Biotechnology, Thycaud Post, Poojappura, Thiruvananthapuram, Trivandrum, 695014 Kerala, India
| | - N K Ganguly
- Institute of Liver and Biliary Science, New Delhi, India
| |
Collapse
|
18
|
Overview of the Development, Impacts, and Challenges of Live-Attenuated Oral Rotavirus Vaccines. Vaccines (Basel) 2020; 8:vaccines8030341. [PMID: 32604982 PMCID: PMC7565912 DOI: 10.3390/vaccines8030341] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Safety, efficacy, and cost-effectiveness are paramount to vaccine development. Following the isolation of rotavirus particles in 1969 and its evidence as an aetiology of severe dehydrating diarrhoea in infants and young children worldwide, the quest to find not only an acceptable and reliable but cost-effective vaccine has continued until now. Four live-attenuated oral rotavirus vaccines (LAORoVs) (Rotarix®, RotaTeq®, Rotavac®, and RotaSIIL®) have been developed and licensed to be used against all forms of rotavirus-associated infection. The efficacy of these vaccines is more obvious in the high-income countries (HIC) compared with the low- to middle-income countries (LMICs); however, the impact is far exceeding in the low-income countries (LICs). Despite the rotavirus vaccine efficacy and effectiveness, more than 90 countries (mostly Asia, America, and Europe) are yet to implement any of these vaccines. Implementation of these vaccines has continued to suffer a setback in these countries due to the vaccine cost, policy, discharging of strategic preventive measures, and infrastructures. This review reappraises the impacts and effectiveness of the current live-attenuated oral rotavirus vaccines from many representative countries of the globe. It examines the problems associated with the low efficacy of these vaccines and the way forward. Lastly, forefront efforts put forward to develop initial procedures for oral rotavirus vaccines were examined and re-connected to today vaccines.
Collapse
|
19
|
Chen YFE, Lee CC, Chiu CH, Chang YC, Tsai CN, Chao HC, Kong SS, Chen SY. Divergence of group a rotavirus with genetic variations before and after introduction of rotavirus vaccines in northern Taiwan. Medicine (Baltimore) 2020; 99:e19253. [PMID: 32118732 PMCID: PMC7478762 DOI: 10.1097/md.0000000000019253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Despite the development of vaccines in 2006, rotavirus is still a major cause of acute gastroenteritis worldwide. This study was performed to analyze the presence of circulating rotaviruses before and after the introduction of rotavirus vaccines to allow phylogenetic comparisons of vaccine strains in northern Taiwan.Rotavirus genotyping and sequencing of rotavirus VP7 and VP4 PCR products were performed by Reverse Transcriptase Polymerase Chain Reaction and DNA autosequencing. Phylogenies were constructed by the neighbor-joining and maximum-likelihood methods using CLUSTAL W software included in the MEGA software package (version 6.0).Between April 2004 and December 2012, a total of 101 rotavirus specimens from pediatric patients with acute gastroenteritis hospitalized in Chang Gung Children's Hospital were amplified, and their VP4 and VP7 sequences were determined. These 101 specimens consisted of 55 pre-vaccine strains (G1 [13, 23.6%], G2 [12, 21.8%], G3 [16, 29.1%], and G9 [14, 25.5%]) and 46 post-vaccine strains (G1 [25, 54.3%], G2 [12, 26.1%], G3 [5, 10.9%], and G9 [4, 8.7%]). The most common combination of the G and P types was G2P[4], accounting for 36% cases, followed by G9P[8] (25%), G1P[8] (20%), G3P[4] (15%), G3P[8] (10%), G1P[4] (5%), and G2P[8] (5%). Phylogenetic analysis showed that only the G1 and P[8] genotypes clustered in the same lineages with the rotavirus vaccine strains.Based on our results, the inclusion of G9, modified G2 and G3 with target lineages, and the combination G2P[4] and G9P[8] in the rotavirus vaccines in Taiwan is warranted as a vaccination strategy.
Collapse
Affiliation(s)
- Ying-Fang Elaine Chen
- Division of Neonatology, Department of Pediatrics, Taipei Medical University Shuang Ho Hospital, New Taipei City
| | - Chung-Chan Lee
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital
| | - Cheng-Hsun Chiu
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine
| | | | - Chi-Neu Tsai
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Department of Pediatrics, Chang Gung Memorial Hospital
| | - Hsun-Ching Chao
- Division of Pediatric Gastroenterology, Chang Gung Children's Hospital, Chang Gung University College of Medicine, Taoyuan
| | | | - Shih-Yen Chen
- Division of Pediatric Gastroenterology, Department of Pediatrics, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan
| |
Collapse
|
20
|
Wagar LE, Bolen CR, Sigal N, Lopez Angel CJ, Guan L, Kirkpatrick BD, Haque R, Tibshirani RJ, Parsonnet J, Petri WA, Davis MM. Increased T Cell Differentiation and Cytolytic Function in Bangladeshi Compared to American Children. Front Immunol 2019; 10:2239. [PMID: 31620139 PMCID: PMC6763580 DOI: 10.3389/fimmu.2019.02239] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 09/04/2019] [Indexed: 01/03/2023] Open
Abstract
During the first 5 years of life, children are especially vulnerable to infection-related morbidity and mortality. Conversely, the Hygiene Hypothesis suggests that a lack of exposure to infectious agents early in life could explain the increasing incidence of allergies and autoimmunity in high-income countries. Understanding these phenomena, however, is hampered by a lack of comprehensive, direct immune monitoring in children with differing degrees of microbial exposure. Using mass cytometry, we provide an in-depth profile of the peripheral blood mononuclear cells (PBMCs) of children in regions at the extremes of exposure: the San Francisco Bay Area, USA and an economically poor district of Dhaka, Bangladesh. Despite variability in clinical health, functional characteristics of PBMCs were similar in Bangladeshi and American children at 1 year of age. However, by 2–3 years of age, Bangladeshi children's immune cells often demonstrated altered activation and cytokine production profiles upon stimulation with PMA-ionomycin, with an overall immune trajectory more in line with American adults. Conversely, immune responses in children from the US remained steady. Using principal component analysis, donor location, ethnic background, and cytomegalovirus infection status were found to account for some of the variation identified among samples. Within Bangladeshi 1-year-olds, stunting (as measured by height-for-age z-scores) was found to be associated with IL-8 and TGFβ expression in PMA-ionomycin stimulated samples. Combined, these findings provide important insights into the immune systems of children in high vs. low microbial exposure environments and suggest an important role for IL-8 and TGFβ in mitigating the microbial challenges faced by the Bangladeshi children.
Collapse
Affiliation(s)
- Lisa E Wagar
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Christopher R Bolen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Natalia Sigal
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, United States
| | - Cesar J Lopez Angel
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Leying Guan
- Data Sciences and Statistics, Stanford University, Stanford, CA, United States
| | - Beth D Kirkpatrick
- Department of Microbiology and Molecular Genetics, University of Vermont College of Medicine and Vaccine Testing Center, Burlington, VT, United States
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Robert J Tibshirani
- Data Sciences and Statistics, Stanford University, Stanford, CA, United States
| | - Julie Parsonnet
- Departments of Medicine and of Health Research and Policy, Stanford University, Stanford, CA, United States
| | - William A Petri
- Department of Medicine, Pathology, and Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States.,Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, United States.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
21
|
Vlasova AN, Takanashi S, Miyazaki A, Rajashekara G, Saif LJ. How the gut microbiome regulates host immune responses to viral vaccines. Curr Opin Virol 2019; 37:16-25. [PMID: 31163292 PMCID: PMC6863389 DOI: 10.1016/j.coviro.2019.05.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 02/07/2023]
Abstract
The co-evolution of the microbiota and immune system has forged a mutually beneficial relationship. This relationship allows the host to maintain the balance between active immunity to pathogens and vaccines and tolerance to self-antigens and food antigens. In children living in low-income and middle-income countries, undernourishment and repetitive gastrointestinal infections are associated with the failure of oral vaccines. Intestinal dysbiosis associated with these environmental influences, as well as some host-related factors, compromises immune responses and negatively impacts vaccine efficacy. To understand how immune responses to viral vaccines can be optimally modulated, mechanistic studies of the relationship between the microbiome, host genetics, viral infections and the development and function of the immune system are needed. We discuss the potential role of the microbiome in modulating vaccine responses in the context of a growing understanding of the relationship between the gastrointestinal microbiota, host related factors (including histo-blood group antigens) and resident immune cell populations.
Collapse
Affiliation(s)
- Anastasia N Vlasova
- Food Animal Health Research Program, CFAES, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA.
| | - Sayaka Takanashi
- Food Animal Health Research Program, CFAES, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA; Department of Developmental Medical Sciences, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ayako Miyazaki
- Division of Viral Disease and Epidemiology, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki 305-0856, Japan
| | - Gireesh Rajashekara
- Food Animal Health Research Program, CFAES, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Linda J Saif
- Food Animal Health Research Program, CFAES, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA.
| |
Collapse
|
22
|
Bucardo F, Reyes Y, Rönnelid Y, González F, Sharma S, Svensson L, Nordgren J. Histo-blood group antigens and rotavirus vaccine shedding in Nicaraguan infants. Sci Rep 2019; 9:10764. [PMID: 31341254 PMCID: PMC6656718 DOI: 10.1038/s41598-019-47166-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022] Open
Abstract
ABO, Lewis and secretor histo-blood group antigens (HBGA) are susceptibility factors for rotavirus in a P-genotype dependent manner and can influence IgA seroconversion rates following rotavirus vaccination. To investigate the association between HBGA phenotypes and rotavirus vaccine shedding fecal samples (n = 304) from a total of 141 infants vaccinated with Rotarix (n = 71) and RotaTeq (n = 70) were prospectively sampled in three time frames (≤3, 4–7 and ≥8 days) after first vaccination dose. Rotavirus was detected with qPCR and genotypes determined by G/P multiplex PCR and/or sequencing. HBGAs were determined by hemagglutination and saliva based ELISA. Low shedding rates were observed, with slightly more children vaccinated with RotaTeq (19%) than Rotarix (11%) shedding rotavirus at ≥4 days post vaccination (DPV). At ≥4 DPV no infant of Lewis A (n = 6) or nonsecretor (n = 9) phenotype in the Rotarix cohort shed rotavirus; the same observation was made for Lewis A infants (n = 7) in the RotaTeq cohort. Putative in-vivo gene reassortment among RotaTeq strains occurred, yielding mainly G1P[8] strains. The bovine derived P[5] genotype included in RotaTeq was able to replicate and be shed at long time frames (>13 DPV). The results of this study are consistent with that HBGA phenotype influences vaccine strain shedding as similarly observed for natural infections. Due to the low overall shedding rates observed, additional studies are however warranted.
Collapse
Affiliation(s)
- Filemón Bucardo
- Department of Microbiology, Faculty of Medical Science, National Autonomous University of Nicaragua, León (UNAN-León), León, Nicaragua.
| | - Yaoska Reyes
- Department of Microbiology, Faculty of Medical Science, National Autonomous University of Nicaragua, León (UNAN-León), León, Nicaragua
| | - Ylva Rönnelid
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Fredman González
- Department of Microbiology, Faculty of Medical Science, National Autonomous University of Nicaragua, León (UNAN-León), León, Nicaragua
| | - Sumit Sharma
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Lennart Svensson
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Johan Nordgren
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| |
Collapse
|
23
|
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]
|
24
|
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
|
25
|
Abebe A, Getahun M, Mapaseka SL, Beyene B, Assefa E, Teshome B, Tefera M, Kebede F, Habtamu A, Haile-Mariam T, Jeffrey Mphahlele M, Teshager F, Ademe A, Teka T, Weldegebriel GG, Mwenda JM. Impact of rotavirus vaccine introduction and genotypic characteristics of rotavirus strains in children less than 5 years of age with gastroenteritis in Ethiopia: 2011-2016. Vaccine 2018; 36:7043-7047. [PMID: 30301641 DOI: 10.1016/j.vaccine.2018.09.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/05/2018] [Accepted: 09/21/2018] [Indexed: 10/28/2022]
Abstract
INTRODUCTION A monovalent rotavirus vaccine was introduced in the Ethiopian Expanded Program on Immunization from November 2013. We compared impact of rotavirus vaccine introduction on rotavirus associated acute diarrhea hospitalizations and genotypic characteristics of rotavirus strains pre-and post-vaccine introduction. METHODS Sentinel surveillance for diarrhea among children <5 years of age was conducted at 3 hospitals in Addis Ababa, Ethiopia from 2011 to 2017. Stool specimens were collected from enrolled children and tested using an antigen capture enzyme immunoassay. Rotavirus positive samples (156 from pre- and 141 from post-vaccination periods) were further characterized by rotavirus genotyping methods to identify the predominant G and P types circulating during the surveillance era. RESULTS A total of 788 children were enrolled during the pre- (July 2011-June 2013) and 815 children during the post-vaccination (July 2014-June 2017) periods. The proportion of diarrhea hospitalizations due to rotavirus among children <5 years of age declined by 17% from 24% (188/788) in the pre-vaccine period and to 20% (161/185) in post-vaccine introduction era. Similarly, a reduction of 18% in proportion of diarrhea hospitalizations due to rotavirus in children <12 months of age in the post (27%) vs pre-vaccine (33%) periods was observed. Seasonal peaks of rotavirus declined following rotavirus vaccine introduction. The most prevalent circulating strains were G12P[8] in 2011 (36%) and in 2012 (27%), G2P[4] (35%) in 2013, G9P[8] (19%) in 2014, G3P[6] and G2P[4] (19% each) in 2015, and G3P[8] (29%) in 2016. DISCUSSION Following rotavirus vaccine introduction in Ethiopia, a reduction in rotavirus associated hospitalizations was seen in all age groups with the greatest burden in children <12 months of age. A wide variety of rotavirus strains circulated in the pre- and post-vaccine introduction periods.
Collapse
Affiliation(s)
- Almaz Abebe
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia.
| | | | - Seheri L Mapaseka
- SAMRC Diarrhoeal Pathogens Research Unit, Department of Virology, Sefako Makgatho Health Sciences University, Medunsa, Pretoria, South Africa
| | - Berhane Beyene
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Essete Assefa
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Birke Teshome
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Mesfin Tefera
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | | | - Abebe Habtamu
- Black Lion Hospital, AAU Medical Faculty, Addis Ababa, Ethiopia
| | | | - M Jeffrey Mphahlele
- SAMRC Diarrhoeal Pathogens Research Unit, Department of Virology, Sefako Makgatho Health Sciences University, Medunsa, Pretoria, South Africa
| | | | | | - Telahun Teka
- Yekatit 12 Hospital, AAU Medical Faculty, Addis Ababa, Ethiopia
| | | | - Jason M Mwenda
- WHO Regional Office for Africa (WHO/AFRO), Brazzaville, People's Republic of Congo
| |
Collapse
|
26
|
Jonesteller CL, Burnett E, Yen C, Tate JE, Parashar UD. Effectiveness of Rotavirus Vaccination: A Systematic Review of the First Decade of Global Postlicensure Data, 2006-2016. Clin Infect Dis 2018; 65:840-850. [PMID: 28444323 DOI: 10.1093/cid/cix369] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/18/2017] [Indexed: 11/13/2022] Open
Abstract
Two rotavirus vaccines, Rotarix (RV1) and RotaTeq (RV5), were licensed for global use in 2006. A systematic review of 48 peer- reviewed articles with postlicensure data from 24 countries showed a median RV1 vaccine effectiveness (VE) of 84%, 75%, and 57% in countries with low, medium, and high child mortality, respectively, and RV5 VE of 90% and 45% in countries with low and high child mortality, respectively. A partial vaccine series provided considerable protection, but not to the same level as a full series. VE tended to decline in the second year of life, particularly in medium- and high-mortality settings, and tended to be greater against more severe rotavirus disease. Postlicensure data from countries across geographic regions and with different child mortality levels demonstrate that under routine use, both RV1 and RV5 are effective against rotavirus disease, supporting the World Health Organization recommendation that all countries introduce rotavirus vaccine into their national immunization program.
Collapse
Affiliation(s)
| | - Eleanor Burnett
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Catherine Yen
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | | |
Collapse
|
27
|
Emergence of Human G2P[4] Rotaviruses in the Post-vaccination Era in South Korea: Footprints of Multiple Interspecies Re-assortment Events. Sci Rep 2018; 8:6011. [PMID: 29662148 PMCID: PMC5902508 DOI: 10.1038/s41598-018-24511-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 04/05/2018] [Indexed: 01/02/2023] Open
Abstract
After the introduction of two global rotavirus vaccines, RotaTeq in 2007 and Rotarix in 2008 in South Korea, G1[P8] rotavirus was the major rotavirus genotype in the country until 2012. However, in this study, an emergence of G2P[4] as the dominant genotype during the 2013 to 2015 season has been reported. Genetic analysis revealed that these viruses had typical DS-1-like genotype constellation and showed evidence of re-assortment in one or more genome segments, including the incorporation of NSP4 genes from strains B-47/2008 from a cow and R4/Haryana/2007 from a buffalo in India, and the VP1 and VP3 genes from strain GO34/1999 from a goat in Bangladesh. Compared to the G2 RotaTeq vaccine strain, 17–24 amino acid changes, specifically A87T, D96N, S213D, and S242N substitutions in G2 epitopes, were observed. These results suggest that multiple interspecies re-assortment events might have contributed to the emergence of G2P[4] rotaviruses in the post-vaccination era in South Korea.
Collapse
|
28
|
Sinha A, Kanungo S, Kim DR, Manna B, Song M, Park JY, Haldar B, Sharma P, Mallick AH, Kim SA, Babji S, Sur D, Kang G, Ali M, Petri WA, Wierzba TF, Czerkinsky C, Nandy RK, Dey A. Antibody secreting B cells and plasma antibody response to rotavirus vaccination in infants from Kolkata India. Heliyon 2018; 4:e00519. [PMID: 29560435 PMCID: PMC5857522 DOI: 10.1016/j.heliyon.2018.e00519] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/09/2018] [Accepted: 01/16/2018] [Indexed: 01/28/2023] Open
Abstract
Background Assessing immune response after rotavirus vaccination consists in measuring serum or plasma IgA and IgG antibodies, but these assays provide very little information about the mucosal immune response. Thus the development of assays for detection of mucosal immune response following rotavirus vaccination is essential. We evaluate to assess circulating antibody-secreting cells (ASCs) as a potential means to evaluate mucosal immune responses to rotavirus vaccine. Methods 372 subjects, aged 6 weeks, were enrolled in the study. All the subjects were assigned to receive two doses of Rotarix® vaccine. Using a micro-modified whole blood-based ELISPOT assay, circulating rotavirus type-specific IgA- and IgG-ASCs, including gut homing β7+ ASCs, were enumerated on week 6 before the first dose of Rotarix vaccination at 7 weeks of age and week 18 after the second vaccination at 17 weeks of age. Plasma samples collected before vaccination, and after two doses of Rotarix® vaccination were tested for plasma rotavirus IgA titers. Results Two doses of Rotarix® provided to induce sero-protective titer of ≥ 20 Units in 35% of subjects. Total blood IgA- ASC responses were detected in 26.4% of subjects who were non-responder before vaccination. Among responders, 47% of the subjects also have sero-protective plasma IgA titers. Discussion Our results suggest that virus-specific blood gut homing ASCs were detected and provide insight into mucosal immune response after rotavirus vaccination. Further studies are needed to evaluate the duration of such immune responses and to assess the programmatic utility of this whole blood-based mucosal ASC testing for the rotavirus immunization program.
Collapse
Affiliation(s)
- Anuradha Sinha
- National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Suman Kanungo
- National Institute of Cholera and Enteric Diseases, Kolkata, India
| | | | - Byomkesh Manna
- National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Manki Song
- International Vaccine Institute, Seoul, South Korea
| | - Ju Yeon Park
- International Vaccine Institute, Seoul, South Korea
| | - Bisakha Haldar
- National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Prashant Sharma
- Department of Microbiology and Immunology, Seoul National University
| | | | - Soon Ae Kim
- International Vaccine Institute, Seoul, South Korea
| | - Sudhir Babji
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Dipika Sur
- National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Gagandeep Kang
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Mohammad Ali
- Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
| | | | | | - Cecil Czerkinsky
- Institut de Pharmacologie Moleculaire & Cellulaire, CNRS-INSERM-University of Nice-Sophia Antipolis, Valbonne, France
| | | | - Ayan Dey
- International Vaccine Institute, Seoul, South Korea
| |
Collapse
|
29
|
The Lewis A phenotype is a restriction factor for Rotateq and Rotarix vaccine-take in Nicaraguan children. Sci Rep 2018; 8:1502. [PMID: 29367698 PMCID: PMC5784145 DOI: 10.1038/s41598-018-19718-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/04/2018] [Indexed: 11/14/2022] Open
Abstract
Histo-blood group antigens (HBGAs) and the Lewis and secretor antigens are associated with susceptibility to rotavirus infection in a genotype-dependent manner. Nicaraguan children were prospectively enrolled in two cohorts vaccinated with either RotaTeq RV5 (n = 68) or Rotarix RV1 (n = 168). Lewis and secretor antigens were determined by saliva phenotyping and genotyping. Seroconversion was defined as a 4-fold increase in plasma IgA antibody titer 1 month after administration of the first dose of the vaccine. Regardless of the vaccine administered, significantly fewer of the children with Lewis A phenotype (0/14) seroconverted after receiving the first vaccine dose compared to 26% (45/175) of those with the Lewis B phenotype and 32% (15/47) of the Lewis negative individuals (P < 0.01). Furthermore, following administration of the RV1 vaccine, secretor-positive ABO blood group B children seroconverted to a significantly lesser extent (5%) compared to secretor-positive children with ABO blood groups A (26%) and O (27%) (P < 0.05). Other factors such as pre-vaccination titers, sex, breastfeeding, and calprotectin levels did not influence vaccine-take. Differences in HBGA expression appear to be a contributing factor in the discrepancy in vaccine-take and thus, in vaccine efficacy in different ethnic populations.
Collapse
|
30
|
|
31
|
Changes in the Occurrence of Rotavirus Gastroenteritis before and after the Introduction of Rotavirus Vaccine among Hospitalized Pediatric Patients and Estimates of Rotavirus Vaccine Effectiveness. ACTA ACUST UNITED AC 2018. [DOI: 10.14776/piv.2018.25.1.26] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
32
|
Affiliation(s)
- Johnathan R Lex
- a College of Medical and Dental Sciences , University of Birmingham , Birmingham , UK
| | | |
Collapse
|
33
|
Park J, Goldstein J, Haran M, Ferrari M. An ensemble approach to predicting the impact of vaccination on rotavirus disease in Niger. Vaccine 2017; 35:5835-5841. [PMID: 28941619 PMCID: PMC7185385 DOI: 10.1016/j.vaccine.2017.09.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/16/2017] [Accepted: 09/05/2017] [Indexed: 01/20/2023]
Abstract
Recently developed vaccines provide a new way of controlling rotavirus in sub-Saharan Africa. Models for the transmission dynamics of rotavirus are critical both for estimating current burden from imperfect surveillance and for assessing potential effects of vaccine intervention strategies. We examine rotavirus infection in the Maradi area in southern Niger using hospital surveillance data provided by Epicentre collected over two years. Additionally, a cluster survey of households in the region allows us to estimate the proportion of children with diarrhea who consulted at a health structure. Model fit and future projections are necessarily particular to a given model; thus, where there are competing models for the underlying epidemiology an ensemble approach can account for that uncertainty. We compare our results across several variants of Susceptible-Infectious-Recovered (SIR) compartmental models to quantify the impact of modeling assumptions on our estimates. Model-specific parameters are estimated by Bayesian inference using Markov chain Monte Carlo. We then use Bayesian model averaging to generate ensemble estimates of the current dynamics, including estimates of R0, the burden of infection in the region, as well as the impact of vaccination on both the short-term dynamics and the long-term reduction of rotavirus incidence under varying levels of coverage. The ensemble of models predicts that the current burden of severe rotavirus disease is 2.6–3.7% of the population each year and that a 2-dose vaccine schedule achieving 70% coverage could reduce burden by 39–42%.
Collapse
Affiliation(s)
- Jaewoo Park
- Department of Statistics, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Joshua Goldstein
- Social and Data Analytics Laboratory, 900 N Glebe Rd, Virginia Tech, Arlington, VA 22203, USA.
| | - Murali Haran
- Department of Statistics, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Matthew Ferrari
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
34
|
Hungerford D, Smith K, Tucker A, Iturriza-Gómara M, Vivancos R, McLeonard C, A Cunliffe N, French N. Population effectiveness of the pentavalent and monovalent rotavirus vaccines: a systematic review and meta-analysis of observational studies. BMC Infect Dis 2017; 17:569. [PMID: 28810833 PMCID: PMC5556361 DOI: 10.1186/s12879-017-2613-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 07/18/2017] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Rotavirus was the leading cause of acute gastroenteritis (AGE) in infants and young children prior to the introduction of routine vaccination. Since 2006 there have been two licensed vaccines available; with successful clinical trials leading the World Health Organization to recommend rotavirus vaccination for all children worldwide. In order to inform immunisation policy we have conducted a systematic review and meta-analysis of observation studies to assess population effectiveness against acute gastroenteritis. METHODS We systematically searched PubMed, Medline, Web of Science, Cinhal and Academic Search Premier and grey literature sources for studies published between January 2006 and April 2014. Studies were eligible for inclusion if they were observational measuring population effectiveness of rotavirus vaccination against health care attendances for rotavirus gastroenteritis or AGE. To evaluate study quality we use used the Newcastle-Ottawa Scale for non-randomised studies, categorising studies by risk of bias. Publication bias was assessed using funnel plots. If two or more studies reported a measure of vaccine effectiveness (VE), we conducted a random effects meta-analysis. We stratified analyses by World Bank country income level and used study quality in sensitivity analyses. RESULTS We identified 30 studies, 19 were from high-income countries and 11 from middle-income countries. Vaccine effectiveness against hospitalization for laboratory confirmed rotavirus gastroenteritis was highest in high-income countries (89% VE; 95% CI 84-92%) compared to middle-income countries (74% VE; 95% CI 67-80%). Vaccine effectiveness was higher for those receiving the complete vaccine schedule (81% VE; 95% CI 75-86%) compared to partial schedule (62% VE; 95% CI 55-69%). Two studies from high-income countries measured VE against community consultations for AGE with a pooled estimate of 40% (95% CI 13-58%; 2 studies). CONCLUSIONS We found strong evidence to further support the continued use of rotavirus vaccines. Vaccine effectiveness was similar to that reported in clinical trials for both high and middle-income countries. There is limited data from Low income settings at present. There was lower effectiveness against milder disease. Further studies, should continue to report effectiveness against AGE and less-severe rotavirus disease because as evidenced by pre-vaccine introduction studies this is likely to contribute the greatest burden on healthcare resources, particularly in high-income countries.
Collapse
Affiliation(s)
- Daniel Hungerford
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK. .,Field Epidemiology Service, National Infection Service, Public Health England, Liverpool, L1 1JF, UK. .,The Centre for Global Vaccine Research, University of Liverpool, Liverpool, L69 7BE, UK.
| | - Katie Smith
- Health Education North West, Liverpool, L3 4BL, UK
| | | | - Miren Iturriza-Gómara
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK.,The Centre for Global Vaccine Research, University of Liverpool, Liverpool, L69 7BE, UK.,NIHR Health Protection Research Unit in Gastrointestinal Infections, Liverpool, UK
| | - Roberto Vivancos
- Field Epidemiology Service, National Infection Service, Public Health England, Liverpool, L1 1JF, UK.,NIHR Health Protection Research Unit in Gastrointestinal Infections, Liverpool, UK.,NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, UK
| | - Catherine McLeonard
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK
| | - Nigel A Cunliffe
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK.,The Centre for Global Vaccine Research, University of Liverpool, Liverpool, L69 7BE, UK.,Department of Medical Microbiology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Neil French
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK.,The Centre for Global Vaccine Research, University of Liverpool, Liverpool, L69 7BE, UK.,Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, UK
| |
Collapse
|
35
|
Verani JR, Baqui AH, Broome CV, Cherian T, Cohen C, Farrar JL, Feikin DR, Groome MJ, Hajjeh RA, Johnson HL, Madhi SA, Mulholland K, O'Brien KL, Parashar UD, Patel MM, Rodrigues LC, Santosham M, Scott JA, Smith PG, Sommerfelt H, Tate JE, Victor JC, Whitney CG, Zaidi AK, Zell ER. Case-control vaccine effectiveness studies: Data collection, analysis and reporting results. Vaccine 2017; 35:3303-3308. [PMID: 28442230 PMCID: PMC7008029 DOI: 10.1016/j.vaccine.2017.04.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/10/2017] [Accepted: 04/12/2017] [Indexed: 12/25/2022]
Abstract
The case-control methodology is frequently used to evaluate vaccine effectiveness post-licensure. The results of such studies provide important insight into the level of protection afforded by vaccines in a 'real world' context, and are commonly used to guide vaccine policy decisions. However, the potential for bias and confounding are important limitations to this method, and the results of a poorly conducted or incorrectly interpreted case-control study can mislead policies. In 2012, a group of experts met to review recent experience with case-control studies evaluating vaccine effectiveness; we summarize the recommendations of that group regarding best practices for data collection, analysis, and presentation of the results of case-control vaccine effectiveness studies. Vaccination status is the primary exposure of interest, but can be challenging to assess accurately and with minimal bias. Investigators should understand factors associated with vaccination as well as the availability of documented vaccination status in the study context; case-control studies may not be a valid method for evaluating vaccine effectiveness in settings where many children lack a documented immunization history. To avoid bias, it is essential to use the same methods and effort gathering vaccination data from cases and controls. Variables that may confound the association between illness and vaccination are also important to capture as completely as possible, and where relevant, adjust for in the analysis according to the analytic plan. In presenting results from case-control vaccine effectiveness studies, investigators should describe enrollment among eligible cases and controls as well as the proportion with no documented vaccine history. Emphasis should be placed on confidence intervals, rather than point estimates, of vaccine effectiveness. Case-control studies are a useful approach for evaluating vaccine effectiveness; however careful attention must be paid to the collection, analysis and presentation of the data in order to best inform evidence-based vaccine policies.
Collapse
Affiliation(s)
- Jennifer R Verani
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA, USA.
| | - Abdullah H Baqui
- International Center for Maternal and Newborn Health, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD, USA
| | - Claire V Broome
- Rollins School of Public Health Emory University, 1518 Clifton Rd, Atlanta, GA, USA
| | - Thomas Cherian
- Department of Immunizations, Vaccines and Biologicals, World Health Organization, 20 Avenue Appia, 1211 Geneva, Switzerland
| | - Cheryl Cohen
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, 1 Modderfontein Rd, Sandringham, Johannesburg, South Africa
| | - Jennifer L Farrar
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA, USA
| | - Daniel R Feikin
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA, USA; International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD, USA
| | - Michelle J Groome
- Respiratory and Meningeal Pathogens Unit, University of Witwatersrand, Richard Ward, 1 Jan Smuts Ave, Braamfontein, Johannesburg, South Africa
| | - Rana A Hajjeh
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA, USA
| | - Hope L Johnson
- Monitoring & Evaluation, Policy & Performance, GAVI Alliance, Chemin des Mines 2, 1202 Geneva, Switzerland
| | - Shabir A Madhi
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, 1 Modderfontein Rd, Sandringham, Johannesburg, South Africa; Respiratory and Meningeal Pathogens Unit, University of Witwatersrand, Richard Ward, 1 Jan Smuts Ave, Braamfontein, Johannesburg, South Africa
| | - Kim Mulholland
- Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville VIC 3052, Australia; Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, Keppel St, London WC1E 7HT, UK
| | - Katherine L O'Brien
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD, USA
| | - Umesh D Parashar
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA, USA
| | - Manish M Patel
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA, USA
| | - Laura C Rodrigues
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, Keppel St, London WC1E 7HT, UK
| | - Mathuram Santosham
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD, USA
| | - J Anthony Scott
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, Keppel St, London WC1E 7HT, UK; KEMRI-Wellcome Trust Research Programme, PO Box 230-80108, Kilifi, Kenya
| | - Peter G Smith
- MRC Tropical Epidemiology Group, London School of Tropical Medicine and Hygiene, London, UK
| | - Halvor Sommerfelt
- Centre of Intervention Science in Maternal and Child Health and Centre for International Health, University of Bergen, PO Box 7800, Bergen, Norway; Department of International Public Health, Norwegian Institute of Public Health, PO Box 4404, Nydalen, Oslo, Norway
| | - Jacqueline E Tate
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA, USA
| | | | - Cynthia G Whitney
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA, USA
| | | | - Elizabeth R Zell
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA, USA
| |
Collapse
|
36
|
Verani JR, Baqui AH, Broome CV, Cherian T, Cohen C, Farrar JL, Feikin DR, Groome MJ, Hajjeh RA, Johnson HL, Madhi SA, Mulholland K, O'Brien KL, Parashar UD, Patel MM, Rodrigues LC, Santosham M, Scott JA, Smith PG, Sommerfelt H, Tate JE, Victor JC, Whitney CG, Zaidi AK, Zell ER. Case-control vaccine effectiveness studies: Preparation, design, and enrollment of cases and controls. Vaccine 2017; 35:3295-3302. [PMID: 28442231 PMCID: PMC7007298 DOI: 10.1016/j.vaccine.2017.04.037] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/10/2017] [Accepted: 04/12/2017] [Indexed: 01/01/2023]
Abstract
Case-control studies are commonly used to evaluate effectiveness of licensed vaccines after deployment in public health programs. Such studies can provide policy-relevant data on vaccine performance under ‘real world’ conditions, contributing to the evidence base to support and sustain introduction of new vaccines. However, case-control studies do not measure the impact of vaccine introduction on disease at a population level, and are subject to bias and confounding, which may lead to inaccurate results that can misinform policy decisions. In 2012, a group of experts met to review recent experience with case-control studies evaluating the effectiveness of several vaccines; here we summarize the recommendations of that group regarding best practices for planning, design and enrollment of cases and controls. Rigorous planning and preparation should focus on understanding the study context including healthcare-seeking and vaccination practices. Case-control vaccine effectiveness studies are best carried out soon after vaccine introduction because high coverage creates strong potential for confounding. Endpoints specific to the vaccine target are preferable to non-specific clinical syndromes since the proportion of non-specific outcomes preventable through vaccination may vary over time and place, leading to potentially confusing results. Controls should be representative of the source population from which cases arise, and are generally recruited from the community or health facilities where cases are enrolled. Matching of controls to cases for potential confounding factors is commonly used, although should be reserved for a limited number of key variables believed to be linked to both vaccination and disease. Case-control vaccine effectiveness studies can provide information useful to guide policy decisions and vaccine development, however rigorous preparation and design is essential.
Collapse
Affiliation(s)
- Jennifer R Verani
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd., Atlanta, GA, USA.
| | - Abdullah H Baqui
- International Center for Maternal and Newborn Health, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD, USA
| | - Claire V Broome
- Rollins School of Public Health Emory University, 1518 Clifton Rd, Atlanta, GA, USA
| | - Thomas Cherian
- Department of Immunizations, Vaccines and Biologicals, World Health Organization, 20 Avenue Appia, 1211 Geneva, Switzerland
| | - Cheryl Cohen
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, 1 Modderfontein Road, Sandringham, Johannesburg, South Africa
| | - Jennifer L Farrar
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd., Atlanta, GA, USA
| | - Daniel R Feikin
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd., Atlanta, GA, USA; International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD, USA
| | - Michelle J Groome
- Respiratory and Meningeal Pathogens Unit, University of Witwatersrand, Richard Ward, 1 Jan Smuts Ave, Braamfontein, Johannesburg, South Africa
| | - Rana A Hajjeh
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd., Atlanta, GA, USA
| | - Hope L Johnson
- Monitoring & Evaluation, Policy & Performance, GAVI Alliance, Chemin des Mines 2, 1202 Geneva, Switzerland
| | - Shabir A Madhi
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, 1 Modderfontein Road, Sandringham, Johannesburg, South Africa; Respiratory and Meningeal Pathogens Unit, University of Witwatersrand, Richard Ward, 1 Jan Smuts Ave, Braamfontein, Johannesburg, South Africa
| | - Kim Mulholland
- Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville, VIC 3052, Australia; Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, Keppel St, London WC1E 7HT, UK
| | - Katherine L O'Brien
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD, USA
| | - Umesh D Parashar
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd., Atlanta, GA, USA
| | - Manish M Patel
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd., Atlanta, GA, USA
| | - Laura C Rodrigues
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, Keppel St, London WC1E 7HT, UK
| | - Mathuram Santosham
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD, USA
| | - J Anthony Scott
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, Keppel St, London WC1E 7HT, UK; KEMRI-Wellcome Trust Research Programme, P.O. Box 230-80108, Kilifi, Kenya
| | - Peter G Smith
- MRC Tropical Epidemiology Group, London School of Tropical Medicine and Hygiene, London, UK
| | - Halvor Sommerfelt
- Centre of Intervention Science in Maternal and Child Health and Centre for International Health, University of Bergen, P.O. Box 7800, Bergen, Norway; Department of International Public Health, Norwegian Institute of Public Health, PO Box 4404, Nydalen, Oslo, Norway
| | - Jacqueline E Tate
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd., Atlanta, GA, USA
| | | | - Cynthia G Whitney
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd., Atlanta, GA, USA
| | | | - Elizabeth R Zell
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd., Atlanta, GA, USA
| |
Collapse
|
37
|
The Association Between Fecal Biomarkers of Environmental Enteropathy and Rotavirus Vaccine Response in Nicaraguan Infants. Pediatr Infect Dis J 2017; 36:412-416. [PMID: 27977553 DOI: 10.1097/inf.0000000000001457] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Environmental enteropathy (EE) is a common intestinal condition among children living in low- and middle-income countries and is associated with diminished enteric immunity to gastrointestinal pathogens, and possibly to oral vaccine antigens. The goal of this study was to examine associations between biomarkers of EE and immunogenicity to the pentavalent rotavirus vaccine (RV5). METHODS Infants were recruited 1 day before their first RV5 immunization in León, Nicaragua, from public health rosters. Infants provided a preimmunization blood and stool sample, and a second blood sample 1 month after receipt of RV5. We measured immunoglobin A (IgA) seroconversion to the first dose of RV5 and concentrations of 4 previously identified fecal biomarkers of EE (alpha-1 antitrypsin, neopterin, myeloperoxidase and calprotectin). We then assessed associations between concentrations of these biomarkers, both individually and as combined scores, and seroconversion to the first dose of RV5. RESULTS Of the 43 enrolled infants, 24 (56%) seroconverted after the first dose of RV5. As compared with infants who seroconverted, those who did not seroconvert had higher median concentrations of both myeloperoxidase (3.1 vs. 1.1 µg/mL, P = 0.002) and calprotectin (199.1 vs. 156.2 µg/mL, P = 0.03). Further, those who did not seroconvert had a higher median combined score of the 4 biomarkers as compared with those who seroconverted (6.5 vs. 4.5, P = 0.017). CONCLUSIONS We found an association between biomarkers of EE and seroconversion to the first dose of RV5. It is possible that interventions that prevent or ameliorate EE may also improve oral rotavirus vaccine response.
Collapse
|
38
|
Luchs A, Timenetsky MDCST. Group A rotavirus gastroenteritis: post-vaccine era, genotypes and zoonotic transmission. EINSTEIN-SAO PAULO 2017; 14:278-87. [PMID: 27462899 PMCID: PMC4943361 DOI: 10.1590/s1679-45082016rb3582] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 02/01/2016] [Indexed: 12/21/2022] Open
Abstract
This article provides a review of immunity, diagnosis, and clinical aspects of rotavirus disease. It also informs about the changes in epidemiology of diarrheal disease and genetic diversity of circulating group A rotavirus strains following the introduction of vaccines. Group A rotavirus is the major pathogen causing gastroenteritis in animals. Its segmented RNA genome can lead to the emergence of new or unusual strains in human populations via interspecies transmission and/or reassortment events.
Collapse
|
39
|
Hull JJ, Cunliffe N, Jere KC, Moon SS, Wang Y, Parashar U, Jiang B. Rotavirus antigen, cytokine, and neutralising antibody profiles in sera of children with and without HIV infection in Blantyre, Malawi. Malawi Med J 2017; 29:24-28. [PMID: 28567192 PMCID: PMC5442487 DOI: 10.4314/mmj.v29i1.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Rotavirus and HIV infection are major causes of death among children in sub-Saharan Africa. A previous study reported no association between concomitant HIV infection and rotavirus disease severity among hospitalised children in Malawi. This study examined rotavirus antigenaemia and broader immune responses among HIV-infected and uninfected children. METHODS Stored (-80°C), paired sera from acute and convalescent phases of Malawian children less than 5 years old, hospitalised for acute gastroenteritis in the primary study, collected from July 1997 to June 1999, were utilised. Among children older than 15 months, HIV infection was defined as the presence of HIV antibody in the blood, when confirmed by at least 2 established methods. For those younger than 15 months, nested polymerase chain reaction (PCR) amplification of proviral DNA was used for verification. All were followed for up to 4 weeks after hospital discharge. Rotavirus antigen levels in sera were measured with Premier™ Rotaclone® rotavirus enzyme immunoassay (EIA) kit. Acute-phase sera were examined for 17 cytokines, using Luminex fluorescent bead human cytokine immunoassay kit. Rotavirus-specific IgA and neutralising activity were determined by EIA and microneutralisation (MN) assay, respectively. Human strains and bovine-human reassortants were propagated in MA104 cells with serum-free Iscove's Modified Dulbecco's Medium (IMDM). Differences in results, from specimens with and without HIV infection, were analysed for statistical significance using the chi-square test. RESULTS We detected rotavirus antigen in 30% of the HIV-infected and 21% HIV-uninfected, in the acute-phase sera. HIV-infected children developed slightly prolonged rotavirus antigenaemia compared to HIV-uninfected children. CONCLUSIONS Rotavirus-specific IgA seroconversion rates and neutralising titres were similar in HIV-infected and HIV-uninfected children, thus, HIV infection had no major effect on immune responses to rotavirus infection.
Collapse
Affiliation(s)
- Jennifer J Hull
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Nigel Cunliffe
- Department of Clinical Infection, Microbiology and Immunology; University of Liverpool, Liverpool, United Kingdom
| | - Khuzwayo C Jere
- Department of Clinical Infection, Microbiology and Immunology; University of Liverpool, Liverpool, United Kingdom
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Sung-Sil Moon
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Yuhuan Wang
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Umesh Parashar
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Baoming Jiang
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
40
|
Becker-Dreps S, Choi WS, Stamper L, Vilchez S, Velasquez DE, Moon SS, Hudgens MG, Jiang B, Permar SR. Innate Immune Factors in Mothers' Breast Milk and Their Lack of Association With Rotavirus Vaccine Immunogenicity in Nicaraguan Infants. J Pediatric Infect Dis Soc 2017; 6:87-90. [PMID: 26582774 PMCID: PMC5907878 DOI: 10.1093/jpids/piv076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 10/13/2015] [Indexed: 12/11/2022]
Abstract
To better understand underlying causes of lower rotavirus vaccine effectiveness in low-middle income countries (LMICs), we measured innate antiviral factors in Nicaraguan mothers' milk and immune response to the first dose of the pentavalent rotavirus vaccine in corresponding infants. No relationship was found between concentrations of innate factors and rotavirus vaccine response.
Collapse
Affiliation(s)
| | - Wan Suk Choi
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
| | - Lisa Stamper
- Department of Pediatrics, Human Vaccine Institute, Duke University, Durham, North Carolina
| | - Samuel Vilchez
- Department of Microbiology and Parasitology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León
| | - Daniel E. Velasquez
- Gastroenteritis and Respiratory Viruses Laboratory Branch, Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Sung-sil Moon
- Gastroenteritis and Respiratory Viruses Laboratory Branch, Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Michael G. Hudgens
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
| | - Baoming Jiang
- Gastroenteritis and Respiratory Viruses Laboratory Branch, Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Sallie R. Permar
- Department of Pediatrics, Human Vaccine Institute, Duke University, Durham, North Carolina
| |
Collapse
|
41
|
Velázquez RF, Linhares AC, Muñoz S, Seron P, Lorca P, DeAntonio R, Ortega-Barria E. Efficacy, safety and effectiveness of licensed rotavirus vaccines: a systematic review and meta-analysis for Latin America and the Caribbean. BMC Pediatr 2017; 17:14. [PMID: 28086819 PMCID: PMC5237165 DOI: 10.1186/s12887-016-0771-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 12/30/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND RotaTeq™ (RV5; Merck & Co. Inc., USA) and Rotarix™ (RV1, GlaxoSmithKline, Belgium) vaccines, developed to prevent rotavirus diarrhea in children under five years old, were both introduced into national immunization programs in 2006. As many countries in Latin America and the Caribbean have included either RV5 or RV1 in their routine childhood vaccination programs, we conducted a systematic review and meta-analysis to analyze efficacy, safety and effectiveness data from the region. METHODS We conducted a systematic search in PubMed, EMBASE, Scielo, Lilacs and the Cochrane Central Register, for controlled efficacy, safety and effectiveness studies published between January 2000 until December 2011, on RV5 and RV1 across Latin America (where both vaccines are available since 2006). The primary outcome measures were: rotavirus-related gastroenteritis of any severity; rotavirus emergency department visits and hospitalization; and severe adverse events. RESULTS The results of the meta-analysis for efficacy show that RV1 reduced the risk of any-severity rotavirus-related gastroenteritis by 65% (relative risk (RR) 0.35, 95% confidence interval (CI) 0.25; 0.50), and of severe gastroenteritis by 82% (RR 0.18, 95%CI 0.12; 0.26) versus placebo. In trials, both vaccines significantly reduced the risk of hospitalization and emergency visits by 85% (RR 0.15, 95%CI 0.09; 0.25) for RV1 and by 90% (RR 0.099, 95%CI 0.012; 0.77) for RV5. Vaccination with RV5 or RV1 did not increase the risk of death, intussusception, or other severe adverse events which were previously associated with the first licensed rotavirus vaccine. Real-world effectiveness studies showed that both vaccines reduced rotavirus hospitalization in the region by around 45-50% for RV5 (for 1 to 3 doses, respectively), and, by around 50-80% for RV1 (for 1 to 2 doses, respectively). For RV1, effectiveness against hospitalization was highest (around 80-96%) for children vaccinated before 12 months of age, compared with 5-60% effectiveness in older children. Both vaccines were most effective in preventing more severe gastroenteritis (70% for RV5 and 80-90% for RV1) and severe gastroenteritis (50% for RV5 and 70-80% for RV1). CONCLUSION This systematic literature review confirms rotavirus vaccination has been proven effective and well tolerated in protecting children in Latin America and the Caribbean.
Collapse
Affiliation(s)
- Raúl F. Velázquez
- Unidad de Investigación Médica en Enfermedades Infecciosas, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Alexandre C. Linhares
- Instituto Evandro Chagas, Secretaria de Vigilância em Saúde, Virology Section, Av. Almirante Barroso 492, 66.090-000 Belém, Pará Brazil
| | - Sergio Muñoz
- Centro de Excelencia Capacitación, Investigación y Gestión para la Salud basada en Evidencias CIGES, Universidad de La Frontera, Temuco, Chile
| | - Pamela Seron
- Centro de Excelencia Capacitación, Investigación y Gestión para la Salud basada en Evidencias CIGES, Universidad de La Frontera, Temuco, Chile
| | - Pedro Lorca
- Centro de Excelencia Capacitación, Investigación y Gestión para la Salud basada en Evidencias CIGES, Universidad de La Frontera, Temuco, Chile
| | | | | |
Collapse
|
42
|
Chen MY, Kirkwood CD, Bines J, Cowley D, Pavlic D, Lee KJ, Orsini F, Watts E, Barnes G, Danchin M. Rotavirus specific maternal antibodies and immune response to RV3-BB neonatal rotavirus vaccine in New Zealand. Hum Vaccin Immunother 2017; 13:1126-1135. [PMID: 28059609 DOI: 10.1080/21645515.2016.1274474] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Maternal antibodies, acquired passively via placenta and/or breast milk, may contribute to the reduced efficacy of oral rotavirus vaccines observed in children in developing countries. This study aimed to investigate the effect of rotavirus specific maternal antibodies on the serum IgA response or stool excretion of vaccine virus after any dose of an oral rotavirus vaccine, RV3-BB, in parallel to a Phase IIa clinical trial conducted at Dunedin Hospital, New Zealand. At the time of the study rotavirus vaccines had not been introduced in New Zealand and the burden of rotavirus disease was evident. METHODS Rotavirus specific IgG and serum neutralizing antibody (SNA) levels in cord blood and IgA levels in colostrum and breast milk samples collected ∼4 weeks, ∼20 weeks and ∼28 weeks after birth were measured. Infants were randomized to receive the first dose of vaccine at 0-5 d (neonatal schedule) or 8 weeks (infant schedule). Breast feeding was with-held for 30 minutes before and after vaccine administration. The relationship between rotavirus specific IgG and SNA levels in cord blood and IgA in colostrum and breast milk at the time of first active dose of RV3-BB vaccine and level of IgA response and stool excretion after 3 doses of vaccine was assessed using linear and logistic regression. RESULTS Forty infants received 3 doses of RV3-BB rotavirus vaccine and were included in the analysis of the neonatal and infant groups. Rotavirus specific IgA in colostrum (neonatal schedule group) and breast milk at 4 weeks (infant schedule group) was identified in 14/21 (67%) and 14/17 (82%) of infants respectively. There was little evidence of an association between IgA in colostrum or breast milk IgA at 4 weeks, or between cord IgG or SNA level, and IgA response or stool excretion after 3 doses of RV3-BB, or after one dose (neonatal schedule) (all p>0.05). CONCLUSIONS The level of IgA in colostrum or breast milk and level of placental IgG and SNA did not impact on the serum IgA response or stool excretion following 3 doses of RV3-BB Rotavirus Vaccine administered using either a neonatal or infant schedule in New Zealand infants.
Collapse
Affiliation(s)
- Mee-Yew Chen
- a Department of Women's and Children's Health , Dunedin School of Medicine, University of Otago , Dunedin , New Zealand
| | - Carl D Kirkwood
- b RV3 Rotavirus Vaccine Program, Murdoch Childrens Research Institute , Parkville , Victoria , Australia.,c Department of Paediatrics , University of Melbourne , Parkville , Victoria , Australia.,e Department of Microbiology , La Trobe University , Bundoora , Victoria , Australia
| | - Julie Bines
- b RV3 Rotavirus Vaccine Program, Murdoch Childrens Research Institute , Parkville , Victoria , Australia.,c Department of Paediatrics , University of Melbourne , Parkville , Victoria , Australia.,d Royal Children's Hospital , Parkville , Victoria , Australia
| | - Daniel Cowley
- b RV3 Rotavirus Vaccine Program, Murdoch Childrens Research Institute , Parkville , Victoria , Australia.,c Department of Paediatrics , University of Melbourne , Parkville , Victoria , Australia
| | - Daniel Pavlic
- b RV3 Rotavirus Vaccine Program, Murdoch Childrens Research Institute , Parkville , Victoria , Australia
| | - Katherine J Lee
- b RV3 Rotavirus Vaccine Program, Murdoch Childrens Research Institute , Parkville , Victoria , Australia.,c Department of Paediatrics , University of Melbourne , Parkville , Victoria , Australia
| | - Francesca Orsini
- b RV3 Rotavirus Vaccine Program, Murdoch Childrens Research Institute , Parkville , Victoria , Australia
| | - Emma Watts
- b RV3 Rotavirus Vaccine Program, Murdoch Childrens Research Institute , Parkville , Victoria , Australia
| | - Graeme Barnes
- b RV3 Rotavirus Vaccine Program, Murdoch Childrens Research Institute , Parkville , Victoria , Australia.,c Department of Paediatrics , University of Melbourne , Parkville , Victoria , Australia
| | - Margaret Danchin
- b RV3 Rotavirus Vaccine Program, Murdoch Childrens Research Institute , Parkville , Victoria , Australia.,c Department of Paediatrics , University of Melbourne , Parkville , Victoria , Australia.,d Royal Children's Hospital , Parkville , Victoria , Australia
| |
Collapse
|
43
|
Leshem E, Givon-Lavi N, Tate JE, Greenberg D, Parashar UD, Dagan R. Real-World Effectiveness of Pentavalent Rotavirus Vaccine Among Bedouin and Jewish Children in Southern Israel. Clin Infect Dis 2016; 62 Suppl 2:S155-60. [PMID: 27059350 DOI: 10.1093/cid/civ1012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Pentavalent rotavirus vaccine (RV5) was introduced into the Israeli National Immunization Program in January 2011. We determined RV5 vaccine effectiveness (VE) in southern Israel, a region characterized by 2 distinct populations: Bedouins living in a low- to middle-income, semirural setting, and Jews living in a high-income, urban setting. METHODS We enrolled vaccine-eligible children who visited the emergency department (ED) or were hospitalized due to acute gastroenteritis (AGE) during the first 3 rotavirus seasons after RV5 vaccine introduction (2011-2013). Fecal specimens were tested for rotavirus by enzyme immunoassay and genotyped. Vaccination among laboratory-confirmed rotavirus cases was compared with rotavirus-negative AGE controls. Regression models were used to calculate VE estimates by age, clinical setting, and ethnicity. RESULTS Of 515 enrolled patients, 359 (70%) were Bedouin. Overall, 185 (36%) patients were rotavirus positive; 79 of 119 (66%) were G1P[8] genotype. The adjusted VE for a full 3-dose course of RV5 against ED visit or hospitalization was 63% (95% confidence interval [CI], 38%-78%). RV5 provided G1P[8] genotype-specific effectiveness of 78% (95% CI, 58%-88%). By age, RV5 VE was 64% (95% CI, 21%-84%) and 71% (95% CI, 39%-86%) among children aged 6-11 months and 12-23 months, respectively. By clinical setting, RV5 VE was 59% (95% CI, 23%-78%) against hospitalization, and 67% (95% CI, 11%-88%) against ED visit. The adjusted VE of a full RV5 course among Bedouin children was 62% (95% CI, 29%-79%). CONCLUSIONS RV5 significantly protected against rotavirus-associated ED visits and hospitalizations in a diverse population of vaccine-eligible children living in southern Israel.
Collapse
Affiliation(s)
- Eyal Leshem
- Internal Medicine C, Sheba Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Israel Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Noga Givon-Lavi
- Pediatric Infectious Diseases Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel
| | - Jacqueline E Tate
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - David Greenberg
- Pediatric Infectious Diseases Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel
| | - Umesh D Parashar
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Ron Dagan
- Pediatric Infectious Diseases Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel
| |
Collapse
|
44
|
Pringle KD, Patzi M, Tate JE, Iniguez Rojas V, Patel M, Inchauste Jordan L, Montesano R, Zarate A, De Oliveira L, Parashar U. Sustained Effectiveness of Rotavirus Vaccine Against Very Severe Rotavirus Disease Through the Second Year of Life, Bolivia 2013-2014. Clin Infect Dis 2016; 62 Suppl 2:S115-20. [PMID: 27059344 DOI: 10.1093/cid/civ1026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND In Bolivia, monovalent rotavirus vaccine was introduced in 2008 and a previous evaluation reported a vaccine effectiveness (VE) of 77% with 2 doses of vaccine in children aged <3 years. This evaluation sought to determine if rotavirus vaccine provided protection through the second year of life against circulating genotypes. METHODS A case-control study was performed in 5 hospitals from April 2013 to March 2014. Among enrolled participants who met study criteria and had rotavirus stool testing performed and vaccine status confirmed, we calculated VE using a logistic regression model. Subgroup analyses were performed among children aged <1 year and those aged ≥1 year, among children with severe diarrhea (Vesikari score ≥11) and very severe diarrhea (Vesikari score ≥15), and among G and P strains with at least 40 specimens. RESULTS A total of 776 children were enrolled. For children <1 year and ≥1 year of age with severe diarrhea, VE for 2 doses was 75% (95% confidence interval [CI], 46%-88%) and 53% (95% CI, 9%-76%), respectively. For children <1 year and ≥1 year of age with very severe diarrhea, VE for 2 doses was 80% (95% CI, 44%-93%) and 74% (95% CI, 35%-90%), respectively. Genotype-specific analysis demonstrated similar VE for the 4 most common G and P types (G3, G9, P[6] and P[8]). CONCLUSIONS A monovalent rotavirus vaccine remains effective against a broad range of circulating strains as part of a routine immunization program >5 years after its introduction in Bolivia. Although VE appears to wane in children aged ≥1 year, it still provides significant protection, and does not wane against severe disease.
Collapse
Affiliation(s)
- Kimberly D Pringle
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases Epidemic Intelligence Service, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Maritza Patzi
- Programa Ampliado de Inmunización, Ministerio de Salud
| | - Jacqueline E Tate
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases
| | - Volga Iniguez Rojas
- Facultad de Ciencias Puras y Naturales, Instituto de Biología Molecular y Biotecnología, Universidad Mayor de San Andrés
| | - Manish Patel
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases
| | - Lucia Inchauste Jordan
- Facultad de Ciencias Puras y Naturales, Instituto de Biología Molecular y Biotecnología, Universidad Mayor de San Andrés
| | | | - Adolfo Zarate
- Programa Ampliado de Inmunización, Ministerio de Salud
| | | | - Umesh Parashar
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases
| |
Collapse
|
45
|
Gastañaduy PA, Steenhoff AP, Mokomane M, Esona MD, Bowen MD, Jibril H, Pernica JM, Mazhani L, Smieja M, Tate JE, Parashar UD, Goldfarb DM. Effectiveness of Monovalent Rotavirus Vaccine After Programmatic Implementation in Botswana: A Multisite Prospective Case-Control Study. Clin Infect Dis 2016; 62 Suppl 2:S161-7. [PMID: 27059351 DOI: 10.1093/cid/civ1207] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Botswana introduced monovalent G1P rotavirus vaccine (RV1) in July 2012, providing one of the first opportunities to assess the effectiveness of routine RV1 vaccination in a high-burden setting in Africa. We sought to determine the effectiveness of RV1 against rotavirus diarrhea hospitalization using a case-control evaluation. METHODS Vaccine age-eligible children <5 years of age admitted with diarrhea at 4 hospitals in Botswana were enrolled from June 2013 to April 2015. Card-confirmed vaccine history was compared between case patients (children with laboratory-confirmed rotavirus diarrhea) and nonrotavirus "test-negative" diarrhea controls. Vaccine effectiveness (VE) was computed using unconditional logistic regression models adjusting for age, birth month/year, and hospital. Sequence-based genotyping was performed on antigen-positive samples. RESULTS Among 242 case patients and 368 controls, 82% (199/242) and 92% (339/368), respectively, had received ≥1 doses of RV1. Effectiveness of a full series (2 doses) of RV1 against rotavirus diarrhea requiring hospitalization was 54% (95% confidence interval [CI], 23%-73%); 1 dose of RV1 was 48% (95% CI, 1%-72%) effective. Effectiveness was 59% (95% CI, 4%-83%) against rotavirus caused by G2P, the most common (37%) circulating genotype. However, the effectiveness of 2 RV1 doses was significantly higher in children with no undernutrition (VE, 75% [95% CI, 41%-89%]), compared to those with moderate or severe undernutrition (VE, -28% [95% CI, -309% to 60%]) (P= .02). CONCLUSIONS Routine RV1 vaccination in Botswana showed effectiveness similar to that in clinical trials in Africa, including against a serotype fully heterotypic to the vaccine. Undernutrition may in part explain the lower rotavirus VE in low-income settings.
Collapse
Affiliation(s)
- Paul A Gastañaduy
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Andrew P Steenhoff
- Department of Paediatrics and Adolescent Health, University of Botswana Botswana-UPenn Partnership, Gaborone Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine
| | - Margaret Mokomane
- Department of Medical Laboratory Sciences, University of Botswana and National Health Laboratory
| | - Mathew D Esona
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Michael D Bowen
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | - Jeffrey M Pernica
- Division of Infectious Disease, Department of Pediatrics, McMaster University, Hamilton, Canada
| | - Loeto Mazhani
- Department of Paediatrics and Adolescent Health, University of Botswana Department of Pediatrics, Princess Marina Hospital, Gaborone, Botswana
| | - Marek Smieja
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton
| | - Jacqueline E Tate
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Umesh D Parashar
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - David M Goldfarb
- Department of Paediatrics and Adolescent Health, University of Botswana Botswana-UPenn Partnership, Gaborone University of British Columbia, Vancouver, Canada
| |
Collapse
|
46
|
Armah G, Pringle K, Enweronu-Laryea CC, Ansong D, Mwenda JM, Diamenu SK, Narh C, Lartey B, Binka F, Grytdal S, Patel M, Parashar U, Lopman B. Impact and Effectiveness of Monovalent Rotavirus Vaccine Against Severe Rotavirus Diarrhea in Ghana. Clin Infect Dis 2016; 62 Suppl 2:S200-7. [PMID: 27059357 DOI: 10.1093/cid/ciw014] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Ghana was among the first African nations to introduce monovalent rotavirus vaccine (RV1) into its childhood immunization schedule in April 2012. We aimed to assess the impact of vaccine introduction on rotavirus and acute gastroenteritis (AGE) hospitalizations and to estimate vaccine effectiveness (VE). METHODS Using data from 2 teaching hospitals, monthly AGE and rotavirus admissions by age were examined 40 months before and 31 months after RV1 introduction using interrupted time-series analyses. From January 2013, we enrolled children <2 years of age who were eligible for RV1 from a total of 7 sentinel sites across the country. To estimate VE, we fit unconditional logistic regression models to calculate odds ratios of vaccination by rotavirus case-patient status, controlling for potential confounders. RESULTS Vaccine coverage ranged from 95% to 100% for dose 1 and 93% to 100% for dose 2. In the first 3 years after vaccine introduction, the percentage of hospital admissions positive for rotavirus fell from 48% in the prevaccine period to 28% (49% adjusted rate reduction; 95% confidence interval [CI], 32%-63%) postvaccination among <5-year-olds. With high vaccine coverage, it was not possible to arrive at robust VE estimates; any-dose VE against rotavirus hospitalization was estimated at 60% (95% CI, -2% to 84%;P= .056). CONCLUSIONS Results from the first 3 years following RV1 introduction suggest substantial reductions of pediatric diarrheal disease as a result of vaccination. Our VE estimate is consistent with the observed rotavirus decrease and with efficacy estimates from elsewhere in sub-Saharan Africa.
Collapse
Affiliation(s)
- George Armah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra
| | | | | | | | - Jason M Mwenda
- World Health Organization Regional Office for Africa, Brazzaville, Republic of Congo
| | | | - Clement Narh
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra School of Public Health, University of Health and Allied Sciences, Hohoe, Ghana
| | - Belinda Lartey
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra
| | - Fred Binka
- School of Public Health, University of Health and Allied Sciences, Hohoe, Ghana
| | - Scott Grytdal
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Manish Patel
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Umesh Parashar
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Ben Lopman
- Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
47
|
Gastañaduy PA, Contreras-Roldán I, Bernart C, López B, Benoit SR, Xuya M, Muñoz F, Desai R, Quaye O, Tam KI, Evans-Bowen DK, Parashar UD, Patel M, McCracken JP. Effectiveness of Monovalent and Pentavalent Rotavirus Vaccines in Guatemala. Clin Infect Dis 2016; 62 Suppl 2:S121-6. [PMID: 27059345 DOI: 10.1093/cid/civ1208] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Concerns remain about lower effectiveness and waning immunity of rotavirus vaccines in resource-poor populations. We assessed vaccine effectiveness against rotavirus in Guatemala, where both the monovalent (RV1; 2-dose series) and pentavalent (RV5; 3-dose series) vaccines were introduced in 2010. METHODS A case-control evaluation was conducted in 4 hospitals from January 2012 to August 2013. Vaccine status was compared between case patients (children with laboratory-confirmed rotavirus diarrhea) and 2 sets of controls: nondiarrhea "hospital" controls (matched by birth date and site) and nonrotavirus "test-negative" diarrhea controls (adjusted for age, birth month/year, and site). Vaccine effectiveness ([1 - odds ratio of vaccination] × 100%) was computed using logistic regression models. RESULTS We evaluated 213 case patients, 657 hospital controls, and 334 test-negative controls. Effectiveness of 2-3 doses of a rotavirus vaccine against rotavirus requiring emergency department visit or hospitalization was 74% (95% confidence interval [CI], 58%-84%) with hospital controls, and 52% (95% CI, 26%-69%) with test-negative controls. Using hospital controls, no significant difference in effectiveness was observed between infants 6-11 months (74% [95% CI, 18%-92%]) and children ≥12 months of age (71% [95% CI, 44%-85%]) (P= .85), nor between complete courses of RV1 (63% [95% CI, 23%-82%]) and RV5 (69% [95% CI, 29%-87%]) (P= .96). An uncommon G12P[8] strain, partially heterotypic to strains in both vaccines, was identified in 89% of cases. CONCLUSIONS RV1 and RV5 were similarly effective against severe rotavirus diarrhea caused by a heterotypic strain in Guatemala. This supports broader implementation of rotavirus vaccination in low-income countries where >90% global deaths from rotavirus occur.
Collapse
Affiliation(s)
- Paul A Gastañaduy
- Epidemic Intelligence Service, Centers for Disease Control and Prevention, Atlanta, Georgia National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | - Chris Bernart
- Center for Health Studies, Universidad del Valle de Guatemala
| | - Beatriz López
- Center for Health Studies, Universidad del Valle de Guatemala
| | - Stephen R Benoit
- International Emerging Infections Program, Centers for Disease Control and Prevention, Guatemala City
| | - Marvin Xuya
- Center for Health Studies, Universidad del Valle de Guatemala
| | - Fredy Muñoz
- Center for Health Studies, Universidad del Valle de Guatemala
| | - Rishi Desai
- Epidemic Intelligence Service, Centers for Disease Control and Prevention, Atlanta, Georgia National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Osbourne Quaye
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia West African Center for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon
| | - Ka Ian Tam
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Diana K Evans-Bowen
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Umesh D Parashar
- Epidemic Intelligence Service, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Manish Patel
- Epidemic Intelligence Service, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | |
Collapse
|
48
|
Hashim ASM, Aboshanab KMA, El-Sayed AFM. Developing an Inactivated Rotavirus Vaccine and Evaluating the Immunogenicity Against a Commercially Available Attenuated Rotavirus Vaccine Using a Mice Animal Model. Viral Immunol 2016; 29:565-571. [PMID: 27860553 DOI: 10.1089/vim.2016.0073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
There is a high demand for public immunization against Rotavirus (RV), especially in Africa. In Africa, the attenuated RV vaccination is contraindicated in patients with immune diseases and nutrition deficiency. Therefore, the inactivated RV vaccine (IRVV) could be an alternative. In this study, we aimed to develop a pentavalent-IRVV using the most circulating RV strains in Egypt and evaluate it against the commercially available Rotarix® vaccine. Trial-IRVV was developed with 5% sucrose, 2% polysorbate-80, and adsorbed on Alum to potentiate the vaccine immune response. Then, it was injected subcutaneously into mice groups at 0-, 21-, and 35-time intervals. In parallel, Rotarix was administered twice on 0 and 28th day. The success of the pentavalent-IRVV/monovalent-Rotarix vaccine immunity rested on achieving immunoglobulin G (IgG) exceeding 1:6,400 that implies less susceptibility to RV infection (RVI). IRVV stimulating IgG >1:6,400 could be an alternative vaccination approach to reach a reasonable protective immunization level against RVI. In addition, Alum adjuvant incorporation effectively provoked a triple elevation of the immunization pattern.
Collapse
Affiliation(s)
- Ayaa S M Hashim
- 1 Research and Development Sector, Egyptian Company for Production of Vaccines, Sera and Drugs-EgyVac (VACSERA Holding Company) , Giza, Egypt
| | - Khaled M A Aboshanab
- 2 Microbiology and Immunology Department, Faculty of Pharmacy, Ain-Shams University , Abbassia, Egypt
| | - Aly F M El-Sayed
- 1 Research and Development Sector, Egyptian Company for Production of Vaccines, Sera and Drugs-EgyVac (VACSERA Holding Company) , Giza, Egypt
| |
Collapse
|
49
|
Gosselin V, Généreux M, Gagneur A, Petit G. Effectiveness of rotavirus vaccine in preventing severe gastroenteritis in young children according to socioeconomic status. Hum Vaccin Immunother 2016; 12:2572-2579. [PMID: 27367155 PMCID: PMC5085015 DOI: 10.1080/21645515.2016.1189038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/24/2016] [Accepted: 05/08/2016] [Indexed: 11/03/2022] Open
Abstract
In 2011, the monovalent rotavirus vaccine was introduced into a universal immunization program in Quebec (Canada). This retrospective cohort study assessed vaccine effectiveness (VE) in preventing acute gastroenteritis (AGE) and rotavirus gastroenteritis (RVGE) hospitalizations among children <3 y living in the Quebec Eastern Townships region according to socioeconomic status (SES). Data were gathered from a tertiary hospital database paired with a regional immunization registry. Three cohorts of children were followed: (1) vaccinated children born in post-universal vaccination period (2011-2013, n = 5,033), (2) unvaccinated children born in post-universal vaccination period (n = 1,239), and (3) unvaccinated children born in pre-universal vaccination period (2008-2010, n = 6,436). In each cohort, AGE and RVGE hospitalizations were identified during equivalent follow-up periods to calculate VE globally and according to neighborhood-level SES. Using multivariable logistic regression, adjusted odds ratios (OR) were computed to obtain VE (1-OR). Adjusted VE of 2 doses was 62% (95% confidence interval [CI]: 37%-77%) and 94% (95%CI: 52%-99%) in preventing AGE and RVGE hospitalization, respectively. Stratified analyses according to SES showed that children living in neighborhoods with higher rates of low-income families had significantly lower VE against AGE hospitalizations compared to neighborhoods with lower rates of low-income families (30% vs. 78%, p = 0.027). Our results suggest that the rotavirus vaccine is highly effective in preventing severe gastroenteritis in young children, particularly among the most well-off. SES seems to influence rotavirus VE, even in a high-income country like Canada. Further studies are needed to determine factors related to lower rotavirus VE among socioeconomically disadvantaged groups.
Collapse
Affiliation(s)
- Virginie Gosselin
- Community Health Sciences Department, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Mélissa Généreux
- Community Health Sciences Department, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
- Eastern Townships Public Health Department, Sherbrooke, Quebec, Canada
| | - Arnaud Gagneur
- Pediatrics Department, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Geneviève Petit
- Community Health Sciences Department, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
- Eastern Townships Public Health Department, Sherbrooke, Quebec, Canada
| |
Collapse
|
50
|
A Systematic Review of the Effect of Rotavirus Vaccination on Diarrhea Outcomes Among Children Younger Than 5 Years. Pediatr Infect Dis J 2016; 35:992-8. [PMID: 27254030 DOI: 10.1097/inf.0000000000001232] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Rotavirus is the leading cause of vaccine-preventable diarrhea among children under 5 globally. Rotavirus vaccination has been shown to prevent severe rotavirus infections with varying efficacy and effectiveness by region. METHODS We sought to generate updated region-specific estimates of rotavirus vaccine efficacy and effectiveness. We systematically reviewed published vaccine efficacy and effectiveness studies to assess the region-specific effect of rotavirus vaccination on select diarrheal morbidity and mortality outcomes in children under 5 years of age. We employed meta-analytic methods to generate pooled effect sizes by Millennium Development Goal region. RESULTS Rotavirus vaccination was both efficacious and effective in preventing rotavirus diarrhea, severe rotavirus diarrhea and rotavirus hospitalizations among children under 5 across all regions represented by the 48 included studies. Efficacy against severe rotavirus diarrhea ranged from 90.6% [95% confidence interval (CI): 82.3-95.0] in the developed region to 88.4% (95% CI: 67.1-95.9) in Eastern/Southeastern Asia, 79.6% (95% CI: 71.3-85.5) in Latin America and the Caribbean, 50.0% (95% CI: 34.4-61.9) in Southern Asia and 46.1% (95% CI: 29.1-59.1) in sub-Saharan Africa. Region-specific effectiveness followed a similar pattern. There was also evidence of vaccine efficacy against severe diarrhea and diarrheal hospitalizations. CONCLUSION Our findings confirm the protective efficacy and effectiveness of rotavirus vaccination against rotavirus diarrheal outcomes among children under 5 globally.
Collapse
|