51
|
Isanaka S, Langendorf C, McNeal MM, Meyer N, Plikaytis B, Garba S, Sayinzoga-Makombe N, Soumana I, Guindo O, Makarimi R, Scherrer MF, Adehossi E, Ciglenecki I, Grais RF. Rotavirus vaccine efficacy up to 2 years of age and against diverse circulating rotavirus strains in Niger: Extended follow-up of a randomized controlled trial. PLoS Med 2021; 18:e1003655. [PMID: 34214095 PMCID: PMC8253401 DOI: 10.1371/journal.pmed.1003655] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/13/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Rotavirus vaccination is recommended in all countries to reduce the burden of diarrhea-related morbidity and mortality in children. In resource-limited settings, rotavirus vaccination in the national immunization program has important cost implications, and evidence for protection beyond the first year of life and against the evolving variety of rotavirus strains is important. We assessed the extended and strain-specific vaccine efficacy of a heat-stable, affordable oral rotavirus vaccine (Rotasiil, Serum Institute of India, Pune, India) against severe rotavirus gastroenteritis (SRVGE) among healthy infants in Niger. METHODS AND FINDINGS From August 2014 to November 2015, infants were randomized in a 1:1 ratio to receive 3 doses of Rotasiil or placebo at approximately 6, 10, and 14 weeks of age. Episodes of gastroenteritis were assessed through active and passive surveillance and graded using the Vesikari score. The primary endpoint was vaccine efficacy of 3 doses of vaccine versus placebo against a first episode of laboratory-confirmed SRVGE (Vesikari score ≥ 11) from 28 days after dose 3, as previously reported. At the time of the primary analysis, median age was 9.8 months. In the present paper, analyses of extended efficacy were undertaken for 3 periods (28 days after dose 3 to 1 year of age, 1 to 2 years of age, and the combined period 28 days after dose 3 to 2 years of age) and by individual rotavirus G type. Among the 3,508 infants included in the per-protocol efficacy analysis (mean age at first dose 6.5 weeks; 49% male), the vaccine provided significant protection against SRVGE through the first year of life (3.96 and 9.98 cases per 100 person-years for vaccine and placebo, respectively; vaccine efficacy 60.3%, 95% CI 43.6% to 72.1%) and over the entire efficacy follow-up period up to 2 years of age (2.13 and 4.69 cases per 100 person-years for vaccine and placebo, respectively; vaccine efficacy 54.7%, 95% CI 38.1% to 66.8%), but the difference was not statistically significant in the second year of life. Up to 2 years of age, rotavirus vaccination prevented 2.56 episodes of SRVGE per 100 child-years. Estimates of efficacy against SRVGE by individual rotavirus genotype were consistent with the overall protective efficacy. Study limitations include limited generalizability to settings with administration of oral polio virus due to low concomitant administration, limited power to assess vaccine efficacy in the second year of life owing to a low number of events among older children, potential bias due to censoring of placebo children at the time of study vaccine receipt, and suboptimal adapted severity scoring based on the Vesikari score, which was designed for use in settings with high parental literacy. CONCLUSIONS Rotasiil provided protection against SRVGE in infants through an extended follow-up period of approximately 2 years. Protection was significant in the first year of life, when the disease burden and risk of death are highest, and against a changing pattern of rotavirus strains during the 2-year efficacy period. Rotavirus vaccines that are safe, effective, and protective against multiple strains represent the best hope for preventing the severe consequences of rotavirus infection, especially in resource-limited settings, where access to care may be limited. Studies such as this provide valuable information for the planning of national immunization programs and future vaccine development. TRIAL REGISTRATION ClinicalTrials.gov NCT02145000.
Collapse
Affiliation(s)
- Sheila Isanaka
- Department of Research, Epicentre, Paris, France
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | | | - Monica Malone McNeal
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Nicole Meyer
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Brian Plikaytis
- BioStat Consulting, Jasper, Georgia, United States of America
| | | | | | | | | | | | | | | | - Iza Ciglenecki
- Operational Center Geneva, Médecins Sans Frontières, Geneva, Switzerland
| | | |
Collapse
|
52
|
Lin FJ, Huang YC, Huang YC, Huang LM, Liu CC, Chi H, Lin HC, Ho YH, Wu FT, Mu JJ, Hsiung CA, Huang CY, Shih SM. Clinical and epidemiological features in hospitalized young children with acute gastroenteritis in Taiwan: A multicentered surveillance through 2014-2017. J Formos Med Assoc 2021; 121:519-528. [PMID: 34167879 DOI: 10.1016/j.jfma.2021.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/20/2021] [Accepted: 06/01/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND/PURPOSE Acute gastroenteritis (AGE) remains a significant health issue in children. The worldwide evolution of pediatric AGE pathogens had been recorded since the introduction of rotavirus vaccine. Ten years after the rotavirus vaccine was introduced to the private sectors in Taiwan, a nationwide study was conducted to elucidate the epidemiological changes among major AGE pathogens. METHODS From January 2014 to December 2017, children younger than 5 years old, hospitalized with AGE at 10 hospitals across Taiwan were enrolled. Stool specimens were tested for Salmonella spp., Campylobacter spp., Clostridiodes difficile, norovirus, and rotavirus by polymerase chain reaction (PCR). The epidemiological and clinical information was collected. RESULTS Enteric pathogen were detected in 1983 (42.2%) of 4700 subjects, with Salmonella spp. (12.5%) being the leading cause of AGE, followed by norovirus (11.2%), rotavirus (8.7%), C. difficile (4.2%), Campylobacter spp. (1.0%), and a mixture of at least 2 of 5 above-mentioned pathogens (4.6%). The case distributions varied across different regions. In eastern Taiwan, rotavirus (21/131, 16.0%) remained the most common pathogen detected. The rotavirus vaccine uptake rate is significantly lower in patients with rotavirus AGE. Besides, rotavirus AGE frequently occurred in children with foreign parent(s), Taiwanese indigenous people, and those with the household monthly income < NT$ 60,000. CONCLUSION Salmonella spp. and norovirus were two major pathogens of pediatric AGE in Taiwan during 2014-17. Providing low-to middle-income households with free rotavirus vaccine nationwide and an industry-led act to reduce salmonellosis should be considered by the authorities.
Collapse
Affiliation(s)
- Fang-Ju Lin
- Division of Infectious Diseases, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Chuan Huang
- Division of Infectious Diseases, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | - Yhu-Chering Huang
- Department of Pediatrics, Chang Gung Children's Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Li-Min Huang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Chuan Liu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsin Chi
- Department of Pediatrics, Mackay Children's Hospital, Mackay Medical College, Taipei, Taiwan
| | - Hsiao-Chuan Lin
- Department of Pediatrics, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Huai Ho
- Division of Infectious Disease, Department of Internal Medicine, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Fang-Tzy Wu
- Center for Diagnostics and Vaccine Development, Centers for Disease Control, Taiwan
| | - Jung-Jung Mu
- Center for Diagnostics and Vaccine Development, Centers for Disease Control, Taiwan
| | - Chao A Hsiung
- Institute of Population Health Sciences, National Health Research Institutes, Taiwan
| | - Ching-Yi Huang
- Institute of Population Health Sciences, National Health Research Institutes, Taiwan
| | - Shu-Man Shih
- Institute of Population Health Sciences, National Health Research Institutes, Taiwan
| | | |
Collapse
|
53
|
Aggarwal S, Hassan E, Baldridge MT. Experimental Methods to Study the Pathogenesis of Human Enteric RNA Viruses. Viruses 2021; 13:975. [PMID: 34070283 PMCID: PMC8225081 DOI: 10.3390/v13060975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/16/2022] Open
Abstract
Every year, millions of children are infected with viruses that target the gastrointestinal tract, causing acute gastroenteritis and diarrheal illness. Indeed, approximately 700 million episodes of diarrhea occur in children under five annually, with RNA viruses norovirus, rotavirus, and astrovirus serving as major causative pathogens. Numerous methodological advancements in recent years, including the establishment of novel cultivation systems using enteroids as well as the development of murine and other animal models of infection, have helped provide insight into many features of viral pathogenesis. However, many aspects of enteric viral infections remain elusive, demanding further study. Here, we describe the different in vitro and in vivo tools available to explore different pathophysiological attributes of human enteric RNA viruses, highlighting their advantages and limitations depending upon the question being explored. In addition, we discuss key areas and opportunities that would benefit from further methodological progress.
Collapse
Affiliation(s)
- Somya Aggarwal
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.A.); (E.H.)
| | - Ebrahim Hassan
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.A.); (E.H.)
| | - Megan T. Baldridge
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.A.); (E.H.)
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
54
|
Gidengil C, Goetz MB, Newberry S, Maglione M, Hall O, Larkin J, Motala A, Hempel S. Safety of vaccines used for routine immunization in the United States: An updated systematic review and meta-analysis. Vaccine 2021; 39:3696-3716. [PMID: 34049735 DOI: 10.1016/j.vaccine.2021.03.079] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Understanding the safety of vaccines is critical to inform decisions about vaccination. Our objective was to conduct a systematic review of the safety of vaccines recommended for children, adults, and pregnant women in the United States. METHODS We searched the literature in November 2020 to update a 2014 Agency for Healthcare Research and Quality review by integrating newly available data. Studies of vaccines that used a comparator and reported the presence or absence of key adverse events were eligible. Adhering to Evidence-based Practice Center methodology, we assessed the strength of evidence (SoE) for all evidence statements. The systematic review is registered in PROSPERO (CRD42020180089). RESULTS Of 56,603 reviewed citations, 338 studies reported in 518 publications met inclusion criteria. For children, SoE was high for no increased risk of autism following measles, mumps, and rubella (MMR) vaccine. SoE was high for increased risk of febrile seizures with MMR. There was no evidence of increased risk of intussusception with rotavirus vaccine at the latest follow-up (moderate SoE), nor of diabetes (high SoE). There was no evidence of increased risk or insufficient evidence for key adverse events for newer vaccines such as 9-valent human papillomavirus and meningococcal B vaccines. For adults, there was no evidence of increased risk (varied SoE) or insufficient evidence for key adverse events for the new adjuvanted inactivated influenza vaccine and recombinant adjuvanted zoster vaccine. We found no evidence of increased risk (varied SoE) for key adverse events among pregnant women following tetanus, diphtheria, and acellular pertussis vaccine, including stillbirth (moderate SoE). CONCLUSIONS Across a large body of research we found few associations of vaccines and serious key adverse events; however, rare events are challenging to study. Any adverse events should be weighed against the protective benefits that vaccines provide.
Collapse
Affiliation(s)
- Courtney Gidengil
- RAND Corporation, 20 Park Plaza, Suite 920, Boston, MA 02116, United States; Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States.
| | - Matthew Bidwell Goetz
- VA Greater Los Angeles Healthcare System and David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90073, United States
| | - Sydne Newberry
- RAND Corporation, 1776 Main Street, Santa Monica, CA 90401, United States
| | - Margaret Maglione
- RAND Corporation, 1776 Main Street, Santa Monica, CA 90401, United States
| | - Owen Hall
- RAND Corporation, 1776 Main Street, Santa Monica, CA 90401, United States
| | - Jody Larkin
- RAND Corporation, 1776 Main Street, Santa Monica, CA 90401, United States
| | - Aneesa Motala
- RAND Corporation, 1776 Main Street, Santa Monica, CA 90401, United States; Southern California Evidence Review Center, University of Southern California, Keck School of Medicine, 2001 N Soto Street, Los Angeles, CA 90033, United States
| | - Susanne Hempel
- RAND Corporation, 1776 Main Street, Santa Monica, CA 90401, United States; Southern California Evidence Review Center, University of Southern California, Keck School of Medicine, 2001 N Soto Street, Los Angeles, CA 90033, United States
| |
Collapse
|
55
|
Hensley C, Zhou P, Schnur S, Mahsoub HM, Liang Y, Wang MX, Page C, Yuan L, Bronshtein V. Thermostable, Dissolvable Buccal Film Rotavirus Vaccine Is Highly Effective in Neonatal Gnotobiotic Pig Challenge Model. Vaccines (Basel) 2021; 9:437. [PMID: 33946555 PMCID: PMC8147248 DOI: 10.3390/vaccines9050437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/27/2021] [Indexed: 01/30/2023] Open
Abstract
Difficulties related to storage and transport of currently available live oral rotavirus vaccines can have detrimental consequences on the efficacy of the vaccines. Thus, there is a great need for thermostable vaccines that can eliminate the necessity for cold chain storage or reconstitution before administration. In this study, we developed a dissolvable oral polymeric film comprised of a live attenuated thermostable tetravalent rhesus-human reassortant rotavirus vaccine (RRV-TV) powder and antacid (CaCO3). Immunogenicity and protective efficacy of the vaccine after buccal delivery was evaluated in the gnotobiotic pig model of human rotavirus (HRV) infection and diarrhea. Two doses of the vaccine were highly immunogenic and conferred strong protection against virus shedding and diarrhea upon challenge with a high dose of a virulent G1 HRV in gnotobiotic pigs. Those pigs vaccinated with the preserved film vaccine had significantly delayed onset of diarrhea; reduced duration and area under the curve of diarrhea; delayed onset of fecal virus shedding; and reduced duration and peak of fecal virus shedding titers compared to pigs in both the placebo and the reconstituted liquid oral RRV-TV vaccine groups. Associated with the strong protection, high titers of serum virus neutralization antibodies against each of the four RRV-TV mono-reassortants and G1 HRV-specific serum IgA and IgG antibodies, as well as intestinal IgA antibodies, were induced by the preserved film vaccine. These results demonstrated the effectiveness of our thermostable buccal film rotavirus vaccine and warrant further investigation into the promise of the novel technology in addressing drawbacks of the current live oral HRV vaccines.
Collapse
Affiliation(s)
- Casey Hensley
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; (C.H.); (P.Z.); (S.S.); (H.M.M.); (Y.L.)
| | - Peng Zhou
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; (C.H.); (P.Z.); (S.S.); (H.M.M.); (Y.L.)
| | - Sofia Schnur
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; (C.H.); (P.Z.); (S.S.); (H.M.M.); (Y.L.)
| | - Hassan M. Mahsoub
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; (C.H.); (P.Z.); (S.S.); (H.M.M.); (Y.L.)
| | - Yu Liang
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; (C.H.); (P.Z.); (S.S.); (H.M.M.); (Y.L.)
| | - Min-Xuan Wang
- Universal Stabilization Technologies, Inc., San Diego, CA 92121, USA; (M.-X.W.); (C.P.)
| | - Caroline Page
- Universal Stabilization Technologies, Inc., San Diego, CA 92121, USA; (M.-X.W.); (C.P.)
| | - Lijuan Yuan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA; (C.H.); (P.Z.); (S.S.); (H.M.M.); (Y.L.)
| | - Victor Bronshtein
- Universal Stabilization Technologies, Inc., San Diego, CA 92121, USA; (M.-X.W.); (C.P.)
| |
Collapse
|
56
|
Abstract
Vaccine efficacy (VE) can vary in different settings. Of the many proposed setting-dependent determinants of VE, force of infection (FoI) stands out as one of the most direct, proximate, and actionable. As highlighted by the COVID-19 pandemic, modifying FoI through non-pharmaceutical interventions (NPIs) use can significantly contribute to controlling transmission and reducing disease incidence and severity absent highly effective pharmaceutical interventions, such as vaccines. Given that NPIs reduce the FoI, the question arises as to if and to what degree FoI, and by extension NPIs, can modify VE, and more practically, as vaccines become available for a pathogen, whether and which NPIs should continue to be used in conjunction with vaccines to optimize controlling transmission and reducing disease incidence and severity.
Collapse
Affiliation(s)
- David C Kaslow
- PATH, 2201 Westlake Avenue, Suite 200, Seattle, WA, USA.
| |
Collapse
|
57
|
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
|
58
|
Wang Y, Li J, Liu P, Zhu F. The performance of licensed rotavirus vaccines and the development of a new generation of rotavirus vaccines: a review. Hum Vaccin Immunother 2021; 17:880-896. [PMID: 32966134 DOI: 10.1080/21645515.2020.1801071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Rotavirus, which causes acute gastroenteritis and severe diarrhea, has posed a great threat to children worldwide over the last 30 y. Since no specific drugs and therapies against rotavirus are available, vaccination is considered the most effective method of decreasing the morbidity and mortality related to rotavirus-associated gastroenteritis. To date, six rotavirus vaccines have been developed and licensed by local governments. Notably, Rotarix™ and RotaTeq™ have been recommended as universal agents against rotavirus infection by the World Health Organization; however, lower efficacies were found in less-developed and developing regions with medium and high child mortality than well-developed ones with low child mortality. For now, two promising novel vaccines, Rotavac™ and RotaSiil™ were pre-qualified by the World Health Organization in 2018. Other rotavirus vaccines in the pipeline including neonatal strain (RV3-BB) and several non-replicating rotavirus vaccines with a parenteral delivery strategy are currently undergoing investigation, with the potential to improve the performance of, and eliminate the safety concerns associated with, previous live oral rotavirus vaccines. This paper reviews the important developments in rotavirus vaccines in the last 20 y and discusses problems and challenges that require investigation in the future.
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
| | - 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
|
59
|
Pradhan SK, Panigrahi S, Padhi PS, Thiyagarajan V, Satpathy SK. Genetic Characteristics of Rotavirus Acute Gastroenteritis Among Hospitalized Children of Odisha in Eastern India. Indian J Pediatr 2021; 88:35-40. [PMID: 33403613 DOI: 10.1007/s12098-020-03610-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/07/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To generate epidemiological data of rotavirus diarrhea among hospitalized children less than 5 y of age and to characterize the circulating rotavirus genotypes post introduction of rotavirus vaccine in Universal Immunization Program (UIP). METHODS This prospective study was conducted from April 2016 to July 2019 at Sardar Vallabhbhai Patel Post Graduate Institute of Paediatrics & SCB Medical College, Cuttack, Odisha among hospitalized children with acute gastroenteritis (AGE) under five years of age. Stool samples collected were tested for rotavirus by a commercial enzyme immunoassay and strains were characterized by reverse-transcription polymerase chain reaction (PCR). The data was analysed using a chi-square test with 95% confidence interval and risk ratio. RESULTS Rotavirus diarrhea was seen in 715 (36.4%) of the 1963 samples tested. The peak incidence of rotavirus diarrhea was during the winter season, i.e., from the month of December to February. Most of the infections were in children between 6 mo to 2 y of age, affecting boys and girls equally. The commonest genotypes were G3P[8] (50.34%) followed by G1P[8] (17.46%). CONCLUSION This study highlights the high prevalence of rotavirus diarrhea among children which emphasize the need for continued rotavirus vaccination. The changing patterns of genotype distribution stress the need for continued surveillance post introduction of vaccines to understand the effect of vaccines on strain evolution over a longer period and detect emergence of new genotypes.
Collapse
Affiliation(s)
- Subal Kumar Pradhan
- Department of Pediatrics, Sardar Vallabhbhai Patel Post Graduate Institute of Paediatrics (SVPPGIP) and SCB Medical College, Cuttack, Odisha, India.
| | - Sumanta Panigrahi
- Department of Pediatrics, Pandit Raghunath Murmu Government Medical College and Hospital, Baripada, Odisha, India
| | - Pooja Sagar Padhi
- Department of Pediatrics, Sardar Vallabhbhai Patel Post Graduate Institute of Paediatrics (SVPPGIP) and SCB Medical College, Cuttack, Odisha, India
| | - Varunkumar Thiyagarajan
- The Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - Saroj Kumar Satpathy
- Department of Pediatrics, Sardar Vallabhbhai Patel Post Graduate Institute of Paediatrics (SVPPGIP) and SCB Medical College, Cuttack, Odisha, India
| |
Collapse
|
60
|
Pasittungkul S, Lestari FB, Puenpa J, Chuchaona W, Posuwan N, Chansaenroj J, Mauleekoonphairoj J, Sudhinaraset N, Wanlapakorn N, Poovorawan Y. High prevalence of circulating DS-1-like human rotavirus A and genotype diversity in children with acute gastroenteritis in Thailand from 2016 to 2019. PeerJ 2021; 9:e10954. [PMID: 33680579 PMCID: PMC7919534 DOI: 10.7717/peerj.10954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/27/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Human rotavirus A (RVA) infection is the primary cause of acute gastroenteritis (AGE) in infants and young children worldwide, especially in children under 5 years of age and is a major public health problem causing severe diarrhea in children in Thailand. This study aimed to investigate the prevalence, genotype diversity, and molecular characterization of rotavirus infection circulating in children under 15 years of age diagnosed with AGE in Thailand from January 2016 to December 2019. METHODS A total of 2,001 stool samples were collected from children with gastroenteritis (neonates to children <15 years of age) and tested for RVA by real-time polymerase chain reaction (RT-PCR). Amplified products were sequenced and submitted to an online genotyping tool for analysis. RESULTS Overall, 301 (15.0%) stool samples were positive for RVA. RVA occurred most frequently among children aged 0-24 months. The seasonal incidence of rotavirus infection occurred typically in Thailand during the winter months (December-March). The G3P[8] genotype was identified as the most prevalent genotype (33.2%, 100/301), followed by G8P[8] (10.6%, 32/301), G9P[8] (6.3%, 19/301), G2P[4] (6.0%, 18/301), and G1P[6] (5.3%, 16/301). Uncommon G and P combinations such as G9P[4], G2P[8], G3P[4] and G3P[9] were also detected at low frequencies. In terms of genetic backbone, the unusual DS-1-like G3P[8] was the most frequently detected (28.2%, 85/301), and the phylogenetic analysis demonstrated high nucleotide identity with unusual DS-1-like G3P[8] detected in Thailand and several countries. CONCLUSIONS A genetic association between RVA isolates from Thailand and other countries ought to be investigated given the local and global dissemination of rotavirus as it is crucial for controlling viral gastroenteritis, and implications for the national vaccination programs.
Collapse
Affiliation(s)
- Siripat Pasittungkul
- Faculty of Medicine, Chulalongkorn University, Center of Excellence in Clinical Virology, Bangkok, Bangkok, Thailand
| | - Fajar Budi Lestari
- Department of Bioresources Technology and Veterinary, Vocational College, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Faculty of Graduate School, Chulalongkorn University, Inter-Department of Biomedical Sciences, Bangkok, Bangkok, Thailand
| | - Jiratchaya Puenpa
- Faculty of Medicine, Chulalongkorn University, Center of Excellence in Clinical Virology, Bangkok, Bangkok, Thailand
| | - Watchaporn Chuchaona
- Faculty of Medicine, Chulalongkorn University, Center of Excellence in Clinical Virology, Bangkok, Bangkok, Thailand
| | - Nawarat Posuwan
- Faculty of Medicine, Chulalongkorn University, Center of Excellence in Clinical Virology, Bangkok, Bangkok, Thailand
| | - Jira Chansaenroj
- Faculty of Medicine, Chulalongkorn University, Center of Excellence in Clinical Virology, Bangkok, Bangkok, Thailand
| | - John Mauleekoonphairoj
- Faculty of Medicine, Chulalongkorn University, Center of Excellence in Clinical Virology, Bangkok, Bangkok, Thailand
| | - Natthinee Sudhinaraset
- Faculty of Medicine, Chulalongkorn University, Center of Excellence in Clinical Virology, Bangkok, Bangkok, Thailand
| | - Nasamon Wanlapakorn
- Faculty of Medicine, Chulalongkorn University, Center of Excellence in Clinical Virology, Bangkok, Bangkok, Thailand
- Faculty of Medicine, Chulalongkorn University, Division of Academic Affairs, Bangkok, Bangkok, Thailand
| | - Yong Poovorawan
- Faculty of Medicine, Chulalongkorn University, Center of Excellence in Clinical Virology, Bangkok, Bangkok, Thailand
| |
Collapse
|
61
|
Baker JM, Tate JE, Leon J, Haber MJ, Pitzer VE, Lopman BA. Postvaccination Serum Antirotavirus Immunoglobulin A as a Correlate of Protection Against Rotavirus Gastroenteritis Across Settings. J Infect Dis 2021; 222:309-318. [PMID: 32060525 DOI: 10.1093/infdis/jiaa068] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 02/13/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND A correlate of protection for rotavirus gastroenteritis would facilitate rapid assessment of vaccination strategies and the next generation of rotavirus vaccines. We aimed to quantify a threshold of postvaccine serum antirotavirus immunoglobulin A (IgA) as an individual-level immune correlate of protection against rotavirus gastroenteritis. METHODS Individual-level data on 5074 infants in 9 GlaxoSmithKline Rotarix Phase 2/3 clinical trials from 16 countries were pooled. Cox proportional hazard models were fit to estimate hazard ratios (HRs) describing the relationship between IgA thresholds and occurrence of rotavirus gastroenteritis. RESULTS Seroconversion (IgA ≥ 20 U/mL) conferred substantial protection against any and severe rotavirus gastroenteritis to age 1 year. In low child mortality settings, seroconversion provided near perfect protection against severe rotavirus gastroenteritis (HR, 0.04; 95% confidence interval [CI], .01-.31). In high child mortality settings, seroconversion dramatically reduced the risk of severe rotavirus gastroenteritis (HR, 0.46; 95% CI, .25-.86). As IgA threshold increased, risk of rotavirus gastroenteritis generally decreased. A given IgA threshold provided better protection in low compared to high child mortality settings. DISCUSSION Postvaccination antirotavirus IgA is a valuable correlate of protection against rotavirus gastroenteritis to age 1 year. Seroconversion provides an informative threshold for assessing rotavirus vaccine performance.
Collapse
Affiliation(s)
- Julia M Baker
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA.,Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jacqueline E Tate
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Juan Leon
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Michael J Haber
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Virginia E Pitzer
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut, USA
| | - Benjamin A Lopman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA.,Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
62
|
Simwaka J, Seheri M, Mulundu G, Kaonga P, Mwenda JM, Chilengi R, Mpabalwani E, Munsaka S. Rotavirus breakthrough infections responsible for gastroenteritis in vaccinated infants who presented with acute diarrhoea at University Teaching Hospitals, Children's Hospital in 2016, in Lusaka Zambia. PLoS One 2021; 16:e0246025. [PMID: 33539399 PMCID: PMC7861525 DOI: 10.1371/journal.pone.0246025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 01/13/2021] [Indexed: 11/19/2022] Open
Abstract
Background In Zambia, before rotavirus vaccine introduction, the virus accounted for about 10 million episodes of diarrhoea, 63 000 hospitalisations and 15 000 deaths in 2015, making diarrhoea the third leading cause of death after pneumonia and malaria. In Zambia, despite the introduction of the vaccine acute diarrhoea due to rotaviruses has continued to affect children aged five years and below. This study aimed to characterise the rotavirus genotypes which were responsible for diarrhoeal infections in vaccinated infants aged 2 to 12 months and to determine the relationship between rotavirus strains and the severity of diarrhoea in 2016. Methods Stool samples from infants aged 2 to 12 months who presented to the hospital with acute diarrhoea of three or more episodes in 24 hours were tested for group A rotavirus. All positive specimens that had enough sample were genotyped using reverse transcriptase Polymerase Chain Reaction (RT-PCR). A 20-point Vesikari clinical score between 1–5 was considered as mild, 6–10 as moderate and greater or equal to 11 as severe. Results A total of 424 stool specimens were tested of which 153 (36%, 95% CI 31.5% to 40.9%) were positive for VP6 rotavirus antigen. The age-specific rotavirus infections decreased significantly (p = 0.041) from 2–4 months, 32.0% (49/118) followed by a 38.8% (70/181) infection rate in the 5–8 months’ category and subsequently dropped in the infants aged 9–12 months with a positivity rate of 27.2%. 38.5% of infants who received a single dose, 34.5% of those who received a complete dose and 45.2% (19/42) of the unvaccinated tested positive for rotavirus. The predominant rotavirus genotypes included G2P[6] 36%, G1P[8] 32%, mixed infections 19%, G2P[4] 6%, G1P[6] 4% and G9P[6] 3%. Discussion and conclusion Results suggest breakthrough infection of heterotypic strains (G2P[6] (36%), homotypic, G1P[8] (32%) and mixed infections (19%) raises concerns about the effects of the vaccination on the rotavirus diversity, considering the selective pressure that rotavirus vaccines could exert on viral populations. This data indicates that the rotavirus vaccine has generally reduced the severity of diarrhoea despite the detection of the virus strains.
Collapse
Affiliation(s)
- Julia Simwaka
- Department of Pathology and Microbiology, School of Medicine, University of Zambia, Lusaka, Zambia
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
- * E-mail:
| | - Mapaseka Seheri
- World Health Organization Regional Office for Africa (WHO/AFRO), Brazzaville, Congo
| | - Gina Mulundu
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
| | - Patrick Kaonga
- Department of Epidemiology and Biostatistics, School of Public Health, University of Zambia, Lusaka, Zambia
- Department of Internal Medicine, University Teaching Hospital, Tropical Gastroenterology and Nutrition Group, Lusaka, Zambia
| | - Jason M. Mwenda
- Department of Virology, Diarrhoea Pathogens Research Unit and WHO AFRO Rotavirus Regional Reference Laboratory, South African Medical Research Council, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| | - Roma Chilengi
- Center for Infectious Disease Research in Zambia, Lusaka, Zambia
| | - Evans Mpabalwani
- Department of Paediatric and Child Health, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Sody Munsaka
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
| |
Collapse
|
63
|
Rakau KG, Nyaga MM, Gededzha MP, Mwenda JM, Mphahlele MJ, Seheri LM, Steele AD. Genetic characterization of G12P[6] and G12P[8] rotavirus strains collected in six African countries between 2010 and 2014. BMC Infect Dis 2021; 21:107. [PMID: 33482744 PMCID: PMC7821174 DOI: 10.1186/s12879-020-05745-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 12/27/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND G12 rotaviruses were first observed in sub-Saharan Africa in 2004 and since then have continued to emerge and spread across the continent and are reported as a significant human rotavirus genotype in several African countries, both prior to and after rotavirus vaccine introduction. This study investigated the genetic variability of 15 G12 rotavirus strains associated with either P[6] or P[8] identified between 2010 and 2014 from Ethiopia, Kenya, Rwanda, Tanzania, Togo and Zambia. METHODS The investigation was carried out by comparing partial VP7 and partial VP4 sequences of the African G12P[6] and G12P[8] strains with the available GenBank sequences and exploring the recognized neutralization epitopes of these strains. Additionally, Bayesian evolutionary analysis was carried out using Markov Chain Monte Carlo (MCMC) implemented in BEAST to estimate the time to the most recent ancestor and evolutionary rate for these G12 rotavirus strains. RESULTS The findings suggested that the VP7 and VP4 nucleotide and amino acid sequences of the G12 strains circulating in African countries are closely related, irrespective of country of origin and year of detection, with the exception of the Ethiopian strains that clustered distinctly. Neutralization epitope analysis revealed that rotavirus VP4 P[8] genes associated with G12 had amino acid sequences similar to those reported globally including the vaccine strains in RotaTeq and Rotarix. The estimated evolutionary rate of the G12 strains was 1.016 × 10- 3 substitutions/site/year and was comparable to what has been previously reported. Three sub-clusters formed within the current circulating lineage III shows the diversification of G12 from three independent ancestries within a similar time frame in the late 1990s. CONCLUSIONS At present it appears to be unlikely that widespread vaccine use has driven the molecular evolution and sustainability of G12 strains in Africa. Continuous monitoring of rotavirus genotypes is recommended to assess the long-term impact of rotavirus vaccination on the dynamic nature of rotavirus evolution on the continent.
Collapse
Affiliation(s)
- Kebareng G Rakau
- Diarrhoeal Pathogens Research Unit, Department of Virology, WHO AFRO Rotavirus Regional Reference Laboratory, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| | - Martin M Nyaga
- Diarrhoeal Pathogens Research Unit, Department of Virology, WHO AFRO Rotavirus Regional Reference Laboratory, Sefako Makgatho Health Sciences University, Pretoria, South Africa.,Next Generation Sequencing Unit and Department of Medical Microbiology and Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Maemu P Gededzha
- Diarrhoeal Pathogens Research Unit, Department of Virology, WHO AFRO Rotavirus Regional Reference Laboratory, Sefako Makgatho Health Sciences University, Pretoria, South Africa.,National Health Laboratory Service, Department of Molecular Medicine and Haematology, Charlotte Maxeke Johannesburg Academic Hospital, Johannesburg, South Africa
| | - Jason M Mwenda
- African Rotavirus Surveillance Network, Immunization, Vaccines and Development Cluster, WHO African Regional Office, Brazzaville, Congo
| | - M Jeffrey Mphahlele
- Diarrhoeal Pathogens Research Unit, Department of Virology, WHO AFRO Rotavirus Regional Reference Laboratory, Sefako Makgatho Health Sciences University, Pretoria, South Africa.,South African Medical Research Council, Soutpansberg Road, Pretoria, South Africa
| | - L Mapaseka Seheri
- Diarrhoeal Pathogens Research Unit, Department of Virology, WHO AFRO Rotavirus Regional Reference Laboratory, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| | - A Duncan Steele
- Diarrhoeal Pathogens Research Unit, Department of Virology, WHO AFRO Rotavirus Regional Reference Laboratory, Sefako Makgatho Health Sciences University, Pretoria, South Africa. .,Present address: Enteric and Diarrheal Diseases, Global Health, Bill & Melinda Gates Foundation, Seattle, WA, USA.
| |
Collapse
|
64
|
Liu C, Huang P, Zhao D, Xia M, Zhong W, Jiang X, Tan M. Effects of rotavirus NSP4 protein on the immune response and protection of the S R69A-VP8* nanoparticle rotavirus vaccine. Vaccine 2021; 39:263-271. [PMID: 33309483 PMCID: PMC7822095 DOI: 10.1016/j.vaccine.2020.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/05/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022]
Abstract
Rotavirus causes severe diarrhea and dehydration in young children. Even with the implementation of the current live vaccines, rotavirus infections still cause significant mortality and morbidity, indicating a need for new rotavirus vaccines with improved efficacy. To this end, we have developed an SR69A-VP8*/S60-VP8* nanoparticle rotavirus vaccine candidate that will be delivered parenterally with Alum adjuvant. In this study, as parts of our further development of this nanoparticle vaccine, we evaluated 1) roles of rotavirus nonstructural protein 4 (NSP4) that is the rotavirus enterotoxin, a possible vaccine target, and an immune stimulator, and 2) effects of CpG adjuvant that is a toll-like receptor 9 (TLR9) ligand and agonist on the immune response and protection of our SR69A-VP8*/S60-VP8* nanoparticle vaccine. The resulted vaccine candidates were examined for their IgG responses in mice. In addition, the resulted mouse sera were assessed for i) blocking titers against interactions of rotavirus VP8* proteins with their glycan ligands, ii) neutralization titers against rotavirus replication in cell culture, and iii) passive protection against rotavirus challenge with diarrhea in suckling mice. Our data showed that the Alum adjuvant appeared to work better with the SR69A-VP8*/S60-VP8* nanoparticles than the CpG adjuvant, while an addition of the NSP4 antigen to the SR69A-VP8*/S60-VP8* vaccine may not help to further increase the immune response and protection of the resulted vaccine.
Collapse
Affiliation(s)
- Cunbao Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China
| | - Pengwei Huang
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Dandan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ming Xia
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Weiming Zhong
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xi Jiang
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ming Tan
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
65
|
Xia M, Huang P, Jiang X, Tan M. A Nanoparticle-Based Trivalent Vaccine Targeting the Glycan Binding VP8* Domains of Rotaviruses. Viruses 2021; 13:72. [PMID: 33419150 PMCID: PMC7825513 DOI: 10.3390/v13010072] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 12/27/2022] Open
Abstract
Rotavirus causes severe gastroenteritis in children. Although vaccines are implemented, rotavirus-related diarrhea still claims ~200,000 lives annually worldwide, mainly in low-income settings, pointing to a need for improved vaccine tactics. To meet such a public health need, a P24-VP8* nanoparticle displaying the glycan-binding VP8* domains, the major neutralizing antigens of rotavirus, was generated as a new type of rotavirus vaccine. We reported here our development of a P24-VP8* nanoparticle-based trivalent vaccine. First, we established a method to produce tag-free P24-VP8* nanoparticles presenting the VP8*s of P[8], P[4], and P[6] rotaviruses, respectively, which are the three predominantly circulating rotavirus P types globally. This approach consists of a chemical-based protein precipitation and an ion exchange purification, which may be scaled up for large vaccine production. All three P24-VP8* nanoparticle types self-assembled efficiently with authentic VP8*-glycan receptor binding function. After they were mixed as a trivalent vaccine, we showed that intramuscular immunization of the vaccine elicited high IgG titers specific to the three homologous VP8* types in mice. The resulted mouse sera strongly neutralized replication of all three rotavirus P types in cell culture. Thus, the trivalent P24-VP8* nanoparticles are a promising vaccine candidate for parenteral use against multiple P types of predominant rotaviruses.
Collapse
Affiliation(s)
- Ming Xia
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (M.X.); (P.H.)
| | - Pengwei Huang
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (M.X.); (P.H.)
| | - Xi Jiang
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (M.X.); (P.H.)
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Ming Tan
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (M.X.); (P.H.)
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
66
|
Esona MD, Gautam R, Katz E, Jaime J, Ward ML, Wikswo ME, Betrapally NS, Rustempasic SM, Selvarangan R, Harrison CJ, Boom JA, Englund J, Klein EJ, Staat MA, McNeal MM, Halasa N, Chappell J, Weinberg GA, Payne DC, Parashar UD, Bowen MD. Comparative genomic analysis of genogroup 1 and genogroup 2 rotaviruses circulating in seven US cities, 2014-2016. Virus Evol 2021; 7:veab023. [PMID: 34522389 PMCID: PMC8432945 DOI: 10.1093/ve/veab023] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
For over a decade, the New Vaccine Surveillance Network (NVSN) has conducted active rotavirus (RVA) strain surveillance in the USA. The evolution of RVA in the post-vaccine introduction era and the possible effects of vaccine pressure on contemporary circulating strains in the USA are still under investigation. Here, we report the whole-gene characterization (eleven ORFs) for 157 RVA strains collected at seven NVSN sites during the 2014 through 2016 seasons. The sequenced strains included 52 G1P[8], 47 G12P[8], 18 G9P[8], 24 G2P[4], 5 G3P[6], as well as 7 vaccine strains, a single mixed strain (G9G12P[8]), and 3 less common strains. The majority of the single and mixed strains possessed a Wa-like backbone with consensus genotype constellation of G1/G3/G9/G12-P[8]-I1-R1-C1-M1-A1-N1-T1-E1-H1, while the G2P[4], G3P[6], and G2P[8] strains displayed a DS-1-like genetic backbone with consensus constellation of G2/G3-P[4]/P[6]/P[8]-I2-R2-C2-M2-A2-N2-T2-E2-H2. Two intergenogroup reassortant G1P[8] strains were detected that appear to be progenies of reassortment events between Wa-like G1P[8] and DS-1-like G2P[4] strains. Two Rotarix® vaccine (RV1) and two RV5 derived (vd) reassortant strains were detected. Phylogenetic and similarity matrices analysis revealed 2-11 sub-genotypic allelic clusters among the genes of Wa- and DS-1-like strains. Most study strains clustered into previously defined alleles. Amino acid (AA) substitutions occurring in the neutralization epitopes of the VP7 and VP4 proteins characterized in this study were mostly neutral in nature, suggesting that these RVA proteins were possibly under strong negative or purifying selection in order to maintain competent and actual functionality, but fourteen radical (AA changes that occur between groups) AA substitutions were noted that may allow RVA strains to gain a selective advantage through immune escape. The tracking of RVA strains at the sub-genotypic allele constellation level will enhance our understanding of RVA evolution under vaccine pressure, help identify possible mechanisms of immune escape, and provide valuable information for formulation of future RVA vaccines.
Collapse
Affiliation(s)
- Mathew D Esona
- Division of Viral Diseases, Centers for Disease Control and Prevention, Viral Gastroenteritis Branch, Atlanta, GA, USA
- Corresponding author: E-mail:
| | - Rashi Gautam
- Division of Viral Diseases, Centers for Disease Control and Prevention, Viral Gastroenteritis Branch, Atlanta, GA, USA
| | - Eric Katz
- Cherokee Nation Assurance, Contracting Agency to the Division of Viral Diseases, Centers for Disease Control and Prevention, Arlington, VA, USA
| | - Jose Jaime
- Division of Viral Diseases, Centers for Disease Control and Prevention, Viral Gastroenteritis Branch, Atlanta, GA, USA
| | - M Leanne Ward
- Division of Viral Diseases, Centers for Disease Control and Prevention, Viral Gastroenteritis Branch, Atlanta, GA, USA
| | - Mary E Wikswo
- Division of Viral Diseases, Centers for Disease Control and Prevention, Viral Gastroenteritis Branch, Atlanta, GA, USA
| | - Naga S Betrapally
- Division of Viral Diseases, Centers for Disease Control and Prevention, Viral Gastroenteritis Branch, Atlanta, GA, USA
| | - Slavica M Rustempasic
- Division of Viral Diseases, Centers for Disease Control and Prevention, Viral Gastroenteritis Branch, Atlanta, GA, USA
| | | | | | | | - Jan Englund
- Seattle Children’s Hospital, Seattle, WA, USA
| | | | - Mary Allen Staat
- Division of Infectious Diseases, Department of Pediatrics, University of Cincinnati, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Monica M McNeal
- Division of Infectious Diseases, Department of Pediatrics, University of Cincinnati, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Natasha Halasa
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - James Chappell
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Geoffrey A Weinberg
- University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Daniel C Payne
- Division of Viral Diseases, Centers for Disease Control and Prevention, Viral Gastroenteritis Branch, Atlanta, GA, USA
| | - Umesh D Parashar
- Division of Viral Diseases, Centers for Disease Control and Prevention, Viral Gastroenteritis Branch, Atlanta, GA, USA
| | - Michael D Bowen
- Division of Viral Diseases, Centers for Disease Control and Prevention, Viral Gastroenteritis Branch, Atlanta, GA, USA
| |
Collapse
|
67
|
Kyo K, Takano C, Kasuga Y, Ogawa E, Ishige M, Pham NTK, Okitsu S, Ushijima H, Urakami T, Fuchigami T, Hayakawa S, Morioka I. Severe rotavirus gastroenteritis in children older than 5 years after vaccine introduction. J Infect Chemother 2020; 27:598-603. [PMID: 33386258 DOI: 10.1016/j.jiac.2020.11.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/29/2020] [Accepted: 11/11/2020] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Rotavirus (RV) is the major pathogen responsible for acute gastroenteritis in infants. Since RV vaccines were introduced, a substantial decline in the incidence of severe RV infection has been reported. However, some burden still exists, even in developed countries, including Japan. METHODS We retrospectively surveyed 380 patients hospitalized for acute gastroenteritis from 2015 to 2019. In 2019, additional detailed clinical information of 21 patients with RV gastroenteritis was obtained to evaluate the efficacy of the RV vaccine. Nine fecal samples from those patients were collected to detect the RV genotypes. RESULTS Our data showed an increasing trend in hospitalizations for severe RV gastroenteritis in children older than 5 years. According to the Vesikari clinical severity scores in the older group (≥5 years), the gastrointestinal symptoms in vaccinated patients were less severe than those in unvaccinated patients (p = 0.014). The genotype analysis revealed that G9P[8]I1 was the major genotype in the recruited patients in 2019. CONCLUSIONS This report warns that children older than 5 years could be affected by severe RV infection and suggests prompt intervention for this age group, similar to that in infants. In the new period in which the RV vaccine is included in Japanese national immunization programs beginning October 2020, continuous monitoring of patient clinical characteristics and RV epidemiology is required to determine the role of vaccines.
Collapse
Affiliation(s)
- Kiyoshi Kyo
- Department of Pediatrics and Child Health, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Chika Takano
- Department of Pediatrics and Child Health, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan; Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan.
| | - Yuki Kasuga
- Department of Pediatrics and Child Health, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Erika Ogawa
- Department of Pediatrics and Child Health, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Mika Ishige
- Department of Pediatrics and Child Health, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Ngan Thi Kim Pham
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Shoko Okitsu
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Hiroshi Ushijima
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Tatsuhiko Urakami
- Department of Pediatrics and Child Health, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Tatsuo Fuchigami
- Department of Pediatrics and Child Health, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan; Department of Pediatrics, IMS Fujimi General Hospital, 1967-1, Tsuruma, Fujimi-shi, Saitama, 354-0021, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Ichiro Morioka
- Department of Pediatrics and Child Health, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| |
Collapse
|
68
|
Standaert B, Strens D, Pereira P, Benninghoff B, Raes M. Lessons Learned from Long-Term Assessment of Rotavirus Vaccination in a High-Income Country: The Case of the Rotavirus Vaccine Belgium Impact Study (RotaBIS). Infect Dis Ther 2020; 9:967-980. [PMID: 33025558 PMCID: PMC7680470 DOI: 10.1007/s40121-020-00345-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/14/2020] [Indexed: 11/28/2022] Open
Abstract
INTRODUCTION The rotavirus (RV) vaccine Belgium Impact Study (RotaBIS) evaluated the vaccine effect on RV-related hospital care in children up to 5 years old over a period of 13 years. Different forces were identified that influence the reduction in hospital care. Our analysis aims to report on the current RotaBIS dataset and explore through model simulation whether, how, and when the results could have been improved. METHODS As performed in previous assessments, this analysis evaluated RV-related events per year, per age group, RV nosocomial infections, hospitalization duration, and herd effect. It subsequently identified results that were surprising or unexpected. To know whether those data could have been improved through specific interventions, we developed a model with the forces acting on the disease transmission and the vaccine effect on RV-related hospital care. Scenario analysis of the forces should explain the current findings and identify ways to optimize the results. RESULTS The RotaBIS data show that annual RV-related hospital cases (n = 1345 pre-vaccination) dropped by 70% (95% confidence interval [CI] 66-74%) by year 5 (n = 395) after vaccine introduction, and by 84% (95% CI 79-89%) by year 10 (n = 217). The herd effect during the first year was limited to 14% extra gain. During the last 5 years, small disease increases were seen biennially. The simulation model indicates that higher vaccine coverage of the major transmitters during the peak season of the first year of vaccination could have reduced RV-related hospital care by nearly 90% at 5 and 10 years after vaccine introduction owing to a higher herd effect. The smaller peaks observed in recent years would have been dramatically reduced. CONCLUSION The current RotaBIS data show a maintained reduction, around 76%, in RV hospitalization cases. Simulations show that these results could have been improved to an important extent with a more optimal initiation of the vaccination program. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT01563146 and NCT01563159.
Collapse
Affiliation(s)
| | | | | | | | - Marc Raes
- Pediatrics, Jessa Ziekenhuis, Hasselt, Belgium
| |
Collapse
|
69
|
Tamim S, Heylen E, Zeller M, Ranst MV, Matthijnssens J, Salman M, Aamir UB, Sharif S, Ikram A, Hasan F. Phylogenetic analysis of open reading frame of 11 gene segments of novel human-bovine reassortant RVA G6P[1] strain in Pakistan. J Med Virol 2020; 92:3179-3186. [PMID: 31696948 DOI: 10.1002/jmv.25625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 10/30/2019] [Indexed: 11/05/2022]
Abstract
Multiple Rotavirus A (RVA) strains are linked with gastrointestinal infections in children that fall in age bracket of 0 to 60 months. However, the problem is augmented with emergence of unique strains that reassort with RVA strains of animal origin. The study describes the sequence analysis of a rare G6P[1] rotavirus strain isolated from a less than 1 year old child, during rotavirus surveillance in Rawalpindi district, Pakistan in 2010. Extracted RNA from fecal specimen was subjected to high throughput RT-PCR for structural and nonstructural gene segments. The complete rotavirus genome of one isolate RVA/Human-wt/PAK/PAK99/2010/G6P[1] was sequenced for phylogenetic analysis to elucidate the evolutionary linkages and origin. Full genome examination of novel strain RVA/Human-wt/PAK/PAK99/2010/G6P[1] revealed the unique genotype assemblage: G6-P[1]-I2-R2-C2-M2-A3-N2-T6-E2-H1. The evolutionary analyses of VP7, VP4, NSP1 and NSP3 gene segments revealed that PAK99 clustered with bovine, or cattle-like rotavirus strains from other closely related species, in the genotypes G6, P[1], A3 and T6 respectively. Gene segments VP6, VP1, VP2, VP3, NSP2 and NSP4 all possessed the DS-1-like bovine genotype 2 and bovine (-like) RVA strains instead of RVA strains having human origin. However, the NSP5 gene was found to cluster closely with contemporary human Wa-like rotavirus strains of H1 genotype. This is the first report on bovine-human (Wa-like reassortant) genotype constellation of G6P[1] strain from a human case in Pakistan (and the second description worldwide). Our results emphasize the significance of incessant monitoring of circulating RVA strains in humans and animals for better understanding of RV evolution.
Collapse
Affiliation(s)
- Sana Tamim
- Public Health Laboratories, Department of Virology/Immunology, National Institute of Health, Islamabad, Pakistan
| | - Elisabeth Heylen
- Laboratory of Virology and Chemotherapy, KU Leuven Department of Microbiology and Immunology, Rega Institute for Medical Research, Leuven, Belgium
| | - Mark Zeller
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California
| | - Marc Van Ranst
- Laboratory for Clinical and Epidemiological Virology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Leuven, Belgium
| | | | - Muhammad Salman
- Public Health Laboratories, Department of Virology/Immunology, National Institute of Health, Islamabad, Pakistan
| | - Uzma Bashir Aamir
- IHP unit Health Emergencies, WHO Country Office, Islamabad, Pakistan
| | - Salman Sharif
- Public Health Laboratories, Department of Virology/Immunology, National Institute of Health, Islamabad, Pakistan
| | - Aamer Ikram
- Public Health Laboratories, Department of Virology/Immunology, National Institute of Health, Islamabad, Pakistan
| | - Fariha Hasan
- Department of Microbiology, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
70
|
Abstract
Enteric viral and bacterial infections continue to be a leading cause of mortality and morbidity in young children in low-income and middle-income countries, the elderly, and immunocompromised individuals. Vaccines are considered an effective and practical preventive approach against the predominantly fecal-to-oral transmitted gastroenteritis particularly in the resource-limited countries or regions where implementation of sanitation systems and supply of safe drinking water are not quickly achievable. While vaccines are available for a few enteric pathogens including rotavirus and cholera, there are no vaccines licensed for many other enteric viral and bacterial pathogens. Challenges in enteric vaccine development include immunological heterogeneity among pathogen strains or isolates, a lack of animal challenge models to evaluate vaccine candidacy, undefined host immune correlates to protection, and a low protective efficacy among young children in endemic regions. In this article, we briefly updated the progress and challenges in vaccines and vaccine development for the leading enteric viral and bacterial pathogens including rotavirus, human calicivirus, Shigella, enterotoxigenic Escherichia coli (ETEC), cholera, nontyphoidal Salmonella, and Campylobacter, and introduced a novel epitope- and structure-based vaccinology platform known as MEFA (multiepitope fusion antigen) and the application of MEFA for developing broadly protective multivalent vaccines against heterogenous pathogens.
Collapse
Affiliation(s)
- Hyesuk Seo
- University of Illinois at Urbana-Champaign, Department of Pathobiology, Urbana, Illinois, USA
| | - Qiangde Duan
- University of Yangzhou, Institute of Comparative Medicine, Yangzhou, PR China
| | - Weiping Zhang
- University of Illinois at Urbana-Champaign, Department of Pathobiology, Urbana, Illinois, USA,CONTACT Weiping Zhang, University of Illinois at Urbana-Champaign, Department of Pathobiology, Urbana, Illinois, USA
| |
Collapse
|
71
|
de Alwis R, My Phuc T, Yu Hang Bai B, Le Thi Quynh N, Thi Thanh Tam P, Thi Ngoc Dung T, Thi Thanh Nhan N, Vinh C, Van Hien H, Thanh Hoang Nhat L, Thi Thu Hong N, Thi Mong Tuyen N, Thi Thuy Trang H, Phuong Thao L, Thi Ngoc Diep V, Thi Hai Chau P, Quan Thinh L, Thi Ngoc Thu H, Nguyet Hang N, Cong Danh M, Doan Hao T, Anh Dao T, Dai L, Thi Huyen Diu V, Thi En N, Thi Tuyet Hanh N, Thi Hanh L, Pham Thu Hien H, Thi Thuy Linh N, Darton TC, Thwaites GE, Kestelyn E, Lan Vi L, Thi Thuy Tien B, Thi Diem Tuyet H, Anderson C, Baker S. The influence of human genetic variation on early transcriptional responses and protective immunity following immunization with Rotarix vaccine in infants in Ho Chi Minh City in Vietnam: A study protocol for an open single-arm interventional trial. Wellcome Open Res 2020. [DOI: 10.12688/wellcomeopenres.16090.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: Rotavirus (RoV) remains the leading cause of acute gastroenteritis in infants and children aged under five years in both high- and low-middle-income countries (LMICs). In LMICs, RoV infections are associated with substantial mortality. Two RoV vaccines (Rotarix and Rotateq) are widely available for use in infants, both of which have been shown to be highly efficacious in Europe and North America. However, for unknown reasons, these RoV vaccines have markedly lower efficacy in LMICs. We hypothesize that poor RoV vaccine efficacy across in certain regions may be associated with genetic heritability or gene expression in the human host. Methods/design: We designed an open-label single-arm interventional trial with the Rotarix RoV vaccine to identify genetic and transcriptomic markers associated with generating a protective immune response against RoV. Overall, 1,000 infants will be recruited prior to Expanded Program on Immunization (EPI) vaccinations at two months of age and vaccinated with oral Rotarix vaccine at two and three months, after which the infants will be followed-up for diarrheal disease until 18 months of age. Blood sampling for genetics, transcriptomics, and immunological analysis will be conducted before each Rotarix vaccination, 2-3 days post-vaccination, and at each follow-up visit (i.e. 6, 12 and 18 months of age). Stool samples will be collected during each diarrheal episode to identify RoV infection. The primary outcome will be Rotarix vaccine failure events (i.e. symptomatic RoV infection despite vaccination), secondary outcomes will be antibody responses and genotypic characterization of the infection virus in Rotarix failure events. Discussion: This study will be the largest and best powered study of its kind to be conducted to date in infants, and will be critical for our understanding of RoV immunity, human genetics in the Vietnam population, and mechanisms determining RoV vaccine-mediated protection. Registration: ClinicalTrials.gov, ID: NCT03587389. Registered on 16 July 2018.
Collapse
|
72
|
Sadiq A, Bostan N. Comparative Analysis of G1P[8] Rotaviruses Identified Prior to Vaccine Implementation in Pakistan With Rotarix™ and RotaTeq™ Vaccine Strains. Front Immunol 2020; 11:562282. [PMID: 33133073 PMCID: PMC7562811 DOI: 10.3389/fimmu.2020.562282] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/21/2020] [Indexed: 01/05/2023] Open
Abstract
Group A rotavirus (RVA) is the leading cause of severe childhood diarrhea globally, even with all effective interventions, particularly in developing countries. Among the diverse genotypes of RVA, G1P[8] is a common genotype that has continued to pervade around the world, including Pakistan. Two universally accepted rotavirus vaccines-Rotarix™ and RotaTeq™ contain the genotype G1P[8]. The current work was aimed at identifying differences between antigenic epitopes of Pakistan’s G1P[8] strains and those of the two licensed vaccines. We sequenced 6 G1P[8] rotavirus strains previously reported in Rawalpindi, Islamabad, Pakistan in 2015 and 2016 for their outer capsid genes (VP7 and VP4). Phylogenetic analysis was then conducted in order to classify their specific lineages and to detect their association with strains isolated throughout world. Compared with the Rotarix™ and RotaTeq™ vaccine strains (G1-lineage II, P[8]-lineage III), our study G1-lineage I, P[8]-lineage IV strains showed 3 and 5 variations in the VP7 epitopes, respectively, and 13 and 11 variations in the VP4 epitopes, respectively. The G1 lineage II strains showed no single amino acid change compared to Rotarix™ (lineage II), but exhibited changes at 2 positions compared to RotaTeq™ (lineage III). So, this has been proposed that these G1 strains exist in our natural setting, or that they may have been introduced in Pakistan from other countries of the world. The distinct P[8]-lineage IV (OP354-like) strains showed twelve and thirteen amino acid variations, with Rotarix™ and RotaTeq™ (lineages II and III) strains, respectively. Such findings have shown that the VP4-P[8] component of the G1P[8] strains circulating in Pakistan differs considerably from that of the vaccine viruses compared to that of the VP7-G1. To monitor the long-term effects of vaccines on the emergence of G1P[8] strains with different lineages, routine and successful monitoring of these strains will be crucial.
Collapse
Affiliation(s)
- Asma Sadiq
- Department of Biosciences, COMSATS University (CUI), Islamabad, Pakistan
| | - Nazish Bostan
- Department of Biosciences, COMSATS University (CUI), Islamabad, Pakistan
| |
Collapse
|
73
|
Kim JH, Kim K, Kim W. Genipin inhibits rotavirus-induced diarrhea by suppressing viral replication and regulating inflammatory responses. Sci Rep 2020; 10:15836. [PMID: 32985574 PMCID: PMC7522720 DOI: 10.1038/s41598-020-72968-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 09/07/2020] [Indexed: 01/16/2023] Open
Abstract
Rotavirus is the leading cause of acute gastroenteritis among young children worldwide. However, agents specifically designed to treat rotavirus infection have not been developed yet. In this study, the anti-rotavirus and anti-inflammatory effects of genipin, a chemical compound found in the fruit of Gardenia jasminoides, were evaluated. Genipin had an antiviral effect against the human rotavirus Wa and SA-11 strains in vitro, and it inhibited two distinct stages of the viral replication cycle: attachment and penetration (early stage) in pre-treatment and assembly and release (late stage) in post-treatment. Additionally, genipin downregulated nitric oxide synthase and pro-inflammatory cytokines in lipopolysaccharide-stimulated RAW264.7 cells and rotavirus-infected Caco-2 cells. Oral administration of genipin before and after viral infection with the murine rotavirus epidemic diarrhea of infant mice strain led to a reduced duration of diarrhea and faecal viral shedding and to decreased destruction of the enteric epithelium. Genipin could have potential as a natural compound with preventive and therapeutic effects against infection and colitis caused by rotavirus.
Collapse
Affiliation(s)
- Jong-Hwa Kim
- Department of Microbiology, Chung-Ang University College of Medicine, 84, Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Kiyoung Kim
- Department of Microbiology, Chung-Ang University College of Medicine, 84, Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Wonyong Kim
- Department of Microbiology, Chung-Ang University College of Medicine, 84, Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea.
| |
Collapse
|
74
|
Babalola MO. Group A Rotavirus G1P[6] Associated Fatalities in Diarrheic Nigerian Infants, Possible Impact of Enterovirus Environmental Enteric Dysfunction, and Implications for Rota-Vaccinology. JOURNAL OF CHILD SCIENCE 2020. [DOI: 10.1055/s-0040-1716374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AbstractGroup A rotavirus (RVA) diarrhea disease and mortality are yet unabated, particularly in developing countries. As global knowledge of specific strains associated with infant mortality is crucial for successful vaccination efforts, candidate RVA strains detected in mortality and fatal cases of severely diarrheic hospitalized infants in Akure, Nigeria were investigated.Fecal samples from comatose patients were tested for RVAs, other diarrhea viruses, and enteric bacterial pathogens. Genomic dsRNA was extracted from 10% rotavirus positive stool suspension, the VP4 and VP7 genes were reverse transcribed and amplified by one-step reverse transcription polymerase chain reaction (PCR) and genotyped by seminested multiplex PCR. Amplicons were sequenced, aligned by ClustalW, and phylogenetic analyses were conducted in MEGA6. Sequences data were deposited to GenBank and DDBJ.Medical examination and microbiological analyses upheld viral diarrhea. EIA revealed RVA and enterovirus. PCR identified virulent RVA strain GIP[6] whose VP7 nucleotide sequences shared a common cluster with Cuban isolate G1P[6], while the VP4 P[6] sequences were related to Asian strains. Reassortant RVA G1P[6] was found in fatal diarrhea cases and mortality of a Nigerian child. RVA coinfection with enterovirus and associated biomarkers of environmental enteric dysfunction in infantile diarrhea should henceforth be evaluated. Current rotavirus vaccines may fare badly against the prevailing virulent strains. The disease severity and outcome necessitates a wider epidemiological study, a review and inclusion of the P[6] genotype in future rotavirus vaccines.
Collapse
Affiliation(s)
- Michael Oluyemi Babalola
- Infectious Diseases Epidemiology, Virology and Special Pathogens Research. Department of Microbiology, Adekunle Ajasin University, Ondo State, Nigeria
- Department of Virology, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan Nigeria
| |
Collapse
|
75
|
Hosomi K, Kunisawa J. Impact of the intestinal environment on the immune responses to vaccination. Vaccine 2020; 38:6959-6965. [PMID: 32933791 DOI: 10.1016/j.vaccine.2020.08.079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/14/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022]
Abstract
Vaccination has contributed greatly to the control of infectious diseases; however, regional and individual differences are occasionally observed in the efficacy of vaccination. As one explanation for these differences, much attention has focused on the intestinal environment constructed by the interaction of diet and the gut microbiota. The intestinal environment has several physiological effects on the host immune system, both locally and systemically, and consequently influences the efficacy of vaccination. In this review, we discuss the impact of the gut microbiota and dietary nutrients on systemic and oral vaccination as well as their applications in various strategies for immunoregulation, including use as vaccine adjuvants. This information could contribute to establishing methods of personalized vaccination that would optimize host immunity by changing the gut environment to maximize vaccine effects.
Collapse
Affiliation(s)
- Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN), Japan.
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN), Japan; International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Japan; Graduate School of Medicine, Graduate School of Pharmaceutical Sciences, Graduate School of Density, Osaka University, Japan; Graduate School of Medicine, Kobe University, Japan; Research Organization for Nano & Life Innovation, Waseda University, Japan.
| |
Collapse
|
76
|
Evaluation of the safety profile of rotavirus vaccines: a pharmacovigilance analysis on American and European data. Sci Rep 2020; 10:13601. [PMID: 32788620 PMCID: PMC7423960 DOI: 10.1038/s41598-020-70653-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 06/08/2020] [Indexed: 11/21/2022] Open
Abstract
Rotaviruses (RVs) are the most common cause of severe diarrheal disease. To date two rotavirus oral vaccines are licensed: Rotarix and Rotateq. Our aim was to contribute to the post-marketing evaluation of these vaccines safety profile. We collected all RV vaccines-related reports of Adverse Events Following Immunization (AEFI) in US Vaccine Adverse Events Reporting System (VAERS) and VigiBase between January 2007 and December 2017. A disproportionality analysis using Reporting Odds Ratio (ROR) was performed. A total of 17,750 reports in VAERS and 6,358 in VigiBase were retrieved. In VAERS, 86.2% of the reports concerned RotaTeq, whereas in VigiBase 67.7% of them involved Rotarix. Across the databases, diarrhea (1,672 events in VAERS, 1,961 in VigiBase) and vomiting (1,746 in VAERS, 1,508 in VigiBase) were the most reported AEFIs. Noteworthy, the RV vaccines-intussusception pair showed a ROR greater than 20 in both databases. Some new potential safety signals emerged such as fontanelle bulging, hypotonic-hyporesponsive episode, livedo reticularis, and opisthotonus. Overall, our data show that most of the reported AEFIs are listed in the Summary of Product Characteristics (SPCs). However, there remains the need to investigate the potential safety signals arose from this analysis, in order to complete the description of the AEFIs.
Collapse
|
77
|
Eraliev U, Latipov R, Tursunova D, Wasley A, Daniels D, Ismoilov U, Akramova M, Sultanova M, Yuldashova D, Barakaev B, Mutalova V, Tuychiev L, Musabaev E, Sharapov S, Pleshkov B, Videbaek D, Huseynov S, Safaeva K, Mijatovic-Rustempasic S, Bowen MD, Parashar UD, Cortese MM. Rotavirus vaccine effectiveness and impact in Uzbekistan, the first country to introduce in central Asia. Hum Vaccin Immunother 2020; 17:503-509. [PMID: 32755429 DOI: 10.1080/21645515.2020.1776034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Uzbekistan, the most populous country in central Asia, was the first in the region to introduce rotavirus vaccine into its national immunization program. Rotarix (GlaxoSmithKline Biologicals, RV1) was introduced in June 2014, with doses recommended at age 2 and 3 months. To evaluate vaccine impact, active surveillance for rotavirus diarrhea was reestablished in 2014 at 2 hospitals in Tashkent and Bukhara which had also performed surveillance during the pre-vaccine period 2005-2009. Children aged <5 y admitted with acute diarrhea had stool specimens collected and tested for rotavirus by enzyme immunoassay. Proportions testing rotavirus-positive in post-vaccine years were compared with the pre-vaccine period. Vaccine records were obtained and effectiveness of 2 RV1 doses vs 0 doses was estimated using rotavirus-case and test-negative design among children enrolled from Bukhara city. In 2015 and 2016, 8%-15% of infants and 10%-16% of children aged<5 y hospitalized with acute diarrhea at the sites tested rotavirus-positive, compared with 26% of infants and 27% of children aged<5 y in pre-vaccine period (reductions in proportion positive of 42%-68%, p <.001). Vaccine effectiveness of 2 RV1 doses vs 0 doses in protecting against hospitalization for rotavirus disease among those aged ≥6 months was 51% (95% CI 2-75) and is based on cases predominantly of genotype G2P[4]. Vaccine effectiveness point estimates tended to be higher against cases with higher illness severity (e.g., clinical severity based on modified Vesikari score ≥11). Our data demonstrate that the monovalent rotavirus vaccine is effective in reducing the likelihood of hospitalization for rotavirus disease in young children in Uzbekistan.
Collapse
Affiliation(s)
| | | | | | - Annemarie Wasley
- World Health Organization Regional Office for Europe , Copenhagen, Denmark
| | - Danni Daniels
- World Health Organization Regional Office for Europe , Copenhagen, Denmark
| | - Umed Ismoilov
- Bukhara Regional Infectious Diseases Hospital , Bukhara, Uzbekistan
| | - Manzura Akramova
- Bukhara Regional Infectious Diseases Hospital , Bukhara, Uzbekistan
| | - Mehri Sultanova
- 4th Tashkent City Infectious Diseases Hospital , Tashkent, Uzbekistan
| | - Dilbar Yuldashova
- 4th Tashkent City Infectious Diseases Hospital , Tashkent, Uzbekistan
| | | | | | | | | | | | - Boris Pleshkov
- Agency for Sanitary-Epidemiological Wellbeing, Ministry of Health , Tashkent, Uzbekistan
| | - Dovile Videbaek
- World Health Organization Regional Office for Europe , Copenhagen, Denmark
| | - Shahin Huseynov
- World Health Organization Regional Office for Europe , Copenhagen, Denmark
| | - Kamola Safaeva
- World Health Organization Country Office in Uzbekistan, Tashkent, Uzbekistan
| | - Slavica Mijatovic-Rustempasic
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, GA, USA
| | - Michael D Bowen
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, GA, USA
| | - Umesh D Parashar
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, GA, USA
| | - Margaret M Cortese
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, GA, USA
| |
Collapse
|
78
|
Pereira P, Vetter V, Standaert B, Benninghoff B. Fifteen years of experience with the oral live-attenuated human rotavirus vaccine: reflections on lessons learned. Expert Rev Vaccines 2020; 19:755-769. [PMID: 32729747 DOI: 10.1080/14760584.2020.1800459] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Rotavirus (RV) disease remains a prominent cause of disease burden in children <5 years of age worldwide. However, implementation of RV vaccination has led to significant reductions in RV mortality, compared to the pre-vaccination era. This review presents 15 years of real-world experience with the oral live-attenuated human RV vaccine (HRV; Rotarix). HRV is currently introduced in ≥80 national immunization programs (NIPs), as 2 doses starting from 6 weeks of age. AREAS COVERED The clinical development of HRV and post-marketing experience indicating the impact of HRV vaccination on RV disease was reviewed. EXPERT OPINION In clinical trials, HRV displayed an acceptable safety profile and efficacy against RV-gastroenteritis, providing broad protection against heterotypic RV strains by reducing the consequences of severe RV disease in infants. Real-world evidence shows substantial, rapid reduction in the number of RV infections and associated hospitalizations following introduction of HRV in NIPs, regardless of economic setting. Indirect effects against RV disease are also observed, such as herd protection, decrease in nosocomial infections incidence, and a reduction of disease-related societal/healthcare costs. However, not all countries have implemented RV vaccination. Coverage remains suboptimal and should be improved to maximize the benefits of RV vaccination.
Collapse
|
79
|
Pitzer VE, Bennett A, Bar-Zeev N, Jere KC, Lopman BA, Lewnard JA, Parashar UD, Cunliffe NA. Evaluating strategies to improve rotavirus vaccine impact during the second year of life in Malawi. Sci Transl Med 2020; 11:11/505/eaav6419. [PMID: 31413144 DOI: 10.1126/scitranslmed.aav6419] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 02/08/2019] [Accepted: 07/25/2019] [Indexed: 01/08/2023]
Abstract
Rotavirus vaccination has substantially reduced the incidence of rotavirus-associated gastroenteritis (RVGE) in high-income countries, but vaccine impact and estimated effectiveness are lower in low-income countries for reasons that are poorly understood. We used mathematical modeling to quantify rotavirus vaccine impact and investigate reduced vaccine effectiveness, particularly during the second year of life, in Malawi, where vaccination was introduced in October 2012 with doses at 6 and 10 weeks. We fitted models to 12 years of prevaccination data and validated the models against postvaccination data to evaluate the magnitude and duration of vaccine protection. The observed rollout of vaccination in Malawi was predicted to lead to a 26 to 77% decrease in the overall incidence of moderate-to-severe RVGE in 2016, depending on assumptions about waning of vaccine-induced immunity and heterogeneity in vaccine response. Vaccine effectiveness estimates were predicted to be higher among 4- to 11-month-olds than 12- to 23-month-olds, even when vaccine-induced immunity did not wane, due to differences in the rate at which vaccinated and unvaccinated individuals acquire immunity from natural infection. We found that vaccine effectiveness during the first and second years of life could potentially be improved by increasing the proportion of infants who respond to vaccination or by lowering the rotavirus transmission rate. An additional dose of rotavirus vaccine at 9 months of age was predicted to lead to higher estimated vaccine effectiveness but to only modest (5 to 16%) reductions in RVGE incidence over the first 3 years after introduction, regardless of assumptions about waning of vaccine-induced immunity.
Collapse
Affiliation(s)
- Virginia E Pitzer
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA.
| | - Aisleen Bennett
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre 3, Malawi.,Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool L69 3BX, UK
| | - Naor Bar-Zeev
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre 3, Malawi.,International Vaccine Access Center, Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Khuzwayo C Jere
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre 3, Malawi.,Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool L69 3BX, UK.,Department of Medical Laboratory Sciences, College of Medicine, University of Malawi, Blantyre 3, Malawi
| | - Benjamin A Lopman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA.,Epidemiology Branch, Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329-4027, USA
| | - Joseph A Lewnard
- Division of Epidemiology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Umesh D Parashar
- Epidemiology Branch, Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329-4027, USA
| | - Nigel A Cunliffe
- Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool L69 3BX, UK
| |
Collapse
|
80
|
Shonhiwa AM, Ntshoe G, Crisp N, Olowolagba AJ, Mbuthu V, Taylor MB, Thomas J, Page NA. Investigation of two suspected diarrhoeal-illness outbreaks in Northern Cape and KwaZulu-Natal provinces, South Africa, April-July 2013: The role of rotavirus. S Afr J Infect Dis 2020; 35:159. [PMID: 34485475 PMCID: PMC8378150 DOI: 10.4102/sajid.v35i1.159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 04/22/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Suspected diarrhoeal-illness outbreaks affecting mostly children < 5 years were investigated between May and July 2013 in Northern Cape province (NCP) and KwaZulu-Natal (KZN) province. This study describes the epidemiological, environmental and clinical characteristics and diarrhoeal-illnesses causative agent(s). METHODS A descriptive cross-sectional study was conducted. Cases were patients presenting at healthcare facilities with diarrhoeal-illness between 09 April and 09 July 2013 in NCP and 01 May and 31 July 2013 in KZN. Laboratory investigations were performed on stools and water samples using microscopy, culture and sensitivity screening and molecular assays. RESULTS A total of 953 cases including six deaths (case fatality rate [CFR]: 0.6%) were recorded in the Northern Cape province outbreak. Children < 5 years accounted for 58% of cases. Enteric viruses were detected in 51% of stools, with rotavirus detected in 43%. The predominant rotavirus strains were G3P[8] (45%) and G9P[8] (42%). Other enteric viruses were detected, with rotavirus co-infections (63%). No enteric pathogens detected in water specimens. KwaZulu-Natal outbreak: A total of 1749 cases including 26 deaths (CFR: 1.5%) were recorded. Children < 5 years accounted for 95% of cases. Rotavirus was detected in 55% of stools; other enteric viruses were detected, mostly as rotavirus co-infections. The predominant rotavirus strains were G2P[4] (54%) and G9P[8] (38%). CONCLUSION Although source(s) of the outbreaks were not identified, the diarrhoeal-illnesses were community-acquired. It is difficult to attribute the outbreaks to one causative agent(s) because of rotavirus co-infections with other enteric pathogens. While rotavirus was predominant, the outbreaks coincided with the annual rotavirus season.
Collapse
Affiliation(s)
- Andronica M Shonhiwa
- Division of Public Health Surveillance and Response, National Institute for Communicable Diseases, National Health Laboratory Service, Sandringham, Johannesburg, South Africa
| | - Genevie Ntshoe
- Division of Public Health Surveillance and Response, National Institute for Communicable Diseases, National Health Laboratory Service, Sandringham, Johannesburg, South Africa
- School of Health Systems and Public Health, Faculty of Health Science, University of Pretoria, Pretoria, South Africa
| | - Noreen Crisp
- Communicable Disease Control, Department of Health, Kimberley, South Africa
| | - Ayo J Olowolagba
- Communicable Disease Control, eThekwini Metropolitan Municipality Department of Health, Durban, South Africa
| | - Vusi Mbuthu
- Communicable Disease Control, eThekwini Metropolitan Municipality Department of Health, Durban, South Africa
| | - Maureen B Taylor
- School of Health Systems and Public Health, Faculty of Health Science, University of Pretoria, Pretoria, South Africa
- Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- National Health Laboratory Service, Tshwane Academic Division, Pretoria, South Africa
| | - Juno Thomas
- Centre for Enteric Diseases, National Institute for Communicable Diseases, National Health Laboratory Service, Sandringham, Johannesburg, South Africa
| | - Nicole A Page
- Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Centre for Enteric Diseases, National Institute for Communicable Diseases, National Health Laboratory Service, Sandringham, Johannesburg, South Africa
| |
Collapse
|
81
|
Impact of Routine Rotavirus Vaccination in Germany: Evaluation Five Years After Its Introduction. Pediatr Infect Dis J 2020; 39:e109-e116. [PMID: 32187139 DOI: 10.1097/inf.0000000000002656] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Routine rotavirus (RV)-vaccination is recommended in Germany since August 2013. Five years later, we evaluated the recommendation by examining vaccine uptake and the impact on RV-gastroenteritis (RVGE) burden in all age groups and on intussusceptions in infants. METHODS We estimated RV-vaccine uptake in the 2014-2018 birth cohorts using statutory health insurance prescription data. For impact assessment, we analyzed RVGE-surveillance data of the German infectious diseases notification system. We compared age-specific RVGE-incidences of different severity between pre-vaccination (2005/06-2007/08) and routine vaccination period (2013/14-2017/18) calculating incidence rate ratios (IRR) using Poisson regression. To determine the effect on intussusception, we used hospital discharge data (2006-2017) and compared incidences between pre-vaccination and routine vaccination period using Poisson regression. RESULTS Vaccination coverage increased from 59% (2014) to 80% (2018). Incidences of RVGE-outpatient cases, RVGE-hospitalization and nosocomial RVGE among <5-year-olds decreased by 74% (IRR = 0.26; 95% CI: 0.26-0.27), 70% (IRR = 0.30; 95% CI: 0.30-0.31) and 70% (IRR = 0.30; 95% CI: 0.30-0.31), respectively. Incidence of RVGE-outpatient cases in age groups ineligible for RV-vaccination decreased by 38% (IRR 0.62; 95% CI: 0.61-0.63). Compared with the pre-vaccination period, incidence of intussusception in the first year of life decreased by 28% (IRR = 0.73; 95% CI: 0.68-0.79) while at age of the first vaccine-dose (7th-12th week of age) increase in incidence of intussusception was non-significant (IRR = 1.29; 95% CI: 0.93-1.78). CONCLUSIONS Routine RV-vaccination is well accepted in Germany. Since implementation of routine RV-vaccination, RVGE significantly decreased in <5-year-olds and in non-vaccinated older age groups through herd protection. The decline of intussusceptions in the first life year suggests a potential vaccination-associated protection against gastrointestinal infections that might trigger intussusceptions. These encouraging results should be communicated to doctors and parents for further improvement of vaccine uptake and protection of more infants.
Collapse
|
82
|
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
|
83
|
De Jesus MCS, Santos VS, Storti-Melo LM, De Souza CDF, Barreto ÍDDC, Paes MVC, Lima PAS, Bohland AK, Berezin EN, Machado RLD, Cuevas LE, Gurgel RQ. Impact of a twelve-year rotavirus vaccine program on acute diarrhea mortality and hospitalization in Brazil: 2006-2018. Expert Rev Vaccines 2020; 19:585-593. [DOI: 10.1080/14760584.2020.1775081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | - Victor Santana Santos
- Departamento De Enfermagem. Núcleo De Epidemiologia E Saúde Pública, Universidade Federal De Alagoas, Arapiraca, Brazil
| | | | | | | | | | | | | | - Eitan N. Berezin
- Department of Pediatrics, Santa Casa De Misericórdia School of Medicine, São Paulo, Brazil
| | - Ricardo Luiz Dantas Machado
- Postgraduate Program in Applied Microbiology and Parasitology, Federal University of Fluminense, Rio De Janeiro, Brazil
- Centro De Investigação De Microrganismos, Universidade Federal Fluminense, Rio De Janeiro, Brazil
| | - Luis Eduardo Cuevas
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | | |
Collapse
|
84
|
Yandle Z, Coughlan S, Dean J, Tuite G, Conroy A, De Gascun CF. Group A Rotavirus Detection and Genotype Distribution before and after Introduction of a National Immunisation Programme in Ireland: 2015-2019. Pathogens 2020; 9:pathogens9060449. [PMID: 32517307 PMCID: PMC7350336 DOI: 10.3390/pathogens9060449] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/05/2020] [Accepted: 06/05/2020] [Indexed: 12/11/2022] Open
Abstract
Immunisation against rotavirus infection was introduced into Ireland in December 2016. We report on the viruses causing gastroenteritis before (2015–2016) and after (2017–2019) implementation of the Rotarix vaccine, as well as changes in the diversity of circulating rotavirus genotypes. Samples from patients aged ≤ 5 years (n = 11,800) were received at the National Virus Reference Laboratory, Dublin, and tested by real-time RT-PCR for rotavirus, Rotarix, norovirus, sapovirus, astrovirus, and enteric adenovirus. Rotavirus genotyping was performed either by multiplex or hemi-nested RT-PCR, and a subset was characterised by sequence analysis. Rotavirus detection decreased by 91% in children aged 0–12 months between 2015/16 and 2018/19. Rotarix was detected in 10% of those eligible for the vaccine and was not found in those aged >7 months. Rotavirus typically peaks in March–May, but following vaccination, the seasonality became less defined. In 2015–16, G1P[8] was the most common genotype circulating; however, in 2019 G2P[4] was detected more often. Following the introduction of Rotarix, a reduction in numbers of rotavirus infections occurred, coinciding with an increase in genotype diversity, along with the first recorded detection of an equine-like G3 strain in Ireland.
Collapse
|
85
|
Asare EO, Al-Mamun MA, Armah GE, Lopman BA, Parashar UD, Binka F, Pitzer VE. Modeling of rotavirus transmission dynamics and impact of vaccination in Ghana. Vaccine 2020; 38:4820-4828. [PMID: 32513513 PMCID: PMC8290434 DOI: 10.1016/j.vaccine.2020.05.057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 04/26/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022]
Abstract
Background: Rotavirus incidence remains relatively high in low-income countries (LICs) compared to high-income countries (HICs) after vaccine introduction. Ghana introduced monovalent rotavirus vaccine in April 2012 and despite the high coverage, vaccine performance has been modest compared to developed countries. The predictors of low vaccine effectiveness in LICs are poorly understood, and the drivers of subnational heterogeneity in rotavirus vaccine impact are unknown. Methods: We used mathematical models to investigate variations in rotavirus incidence in children <5 years old in Ghana. We fit models to surveillance and case-control data from three different hospitals: Korle-Bu Teaching Hospital in Accra, Komfo Anokye Teaching Hospital in Kumasi, and War Memorial Hospital in Navrongo. The models were fitted to both pre- and post-vaccine data to estimate parameters describing the transmission rate, waning of maternal immunity, and vaccine response rate. Results: The seasonal pattern and age distribution of rotavirus cases varied among the three study sites in Ghana. Our model was able to capture the spatio-temporal variations in rotavirus incidence across the three sites and showed good agreement with the age distribution of observed cases. The rotavirus transmission rate was highest in Accra and lowest in Navrongo, while the estimated duration of maternal immunity was longer (∼5 months) in Accra and Kumasi and shorter (∼3 months) in Navrongo. The proportion of infants who responded to the vaccine was estimated to be high in Accra and Kumasi and low in Navrongo. Conclusions: Rotavirus vaccine impact varies within Ghana. A low vaccine response rate was estimated for Navrongo, where rotavirus is highly seasonal and incidence limited to a few months of the year. Our findings highlight the need to further explore the relationship between rotavirus seasonality, maternal immunity, and vaccine response rate to determine how they influence vaccine effectiveness and to develop strategies to improve vaccine impact.
Collapse
Affiliation(s)
- Ernest O Asare
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT, USA.
| | - Mohammad A Al-Mamun
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - George E Armah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Benjamin A Lopman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Umesh D Parashar
- Epidemiology Branch, Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Fred Binka
- University of Health and Allied Health Sciences, Ho, Ghana
| | - Virginia E Pitzer
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT, USA
| |
Collapse
|
86
|
Phylodynamics of G4P[8] and G2P[4] strains of rotavirus A isolated in Russia in 2017 based on full-genome analyses. Virus Genes 2020; 56:537-545. [PMID: 32472472 DOI: 10.1007/s11262-020-01771-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/22/2020] [Indexed: 01/08/2023]
Abstract
Rotavirus A is a dynamically evolving pathogen causing acute gastroenteritis in children during the first years of life. In the present study, we conducted a phylodynamic analysis based on the complete sequences of 11 segments of rotaviruses with the G4P[8] and G2P[4] genotypes isolated in Russia in 2017. Since rotavirus has a segmented genome, our analysis was performed using the Bayesian approach based on separate samples of nucleotide sequences for each gene of the strains studied. For the strain with the genotype G4P[8], the most likely geographical locations of the nearest common ancestor were Russia (VP7, VP4, VP6), China (VP1), Thailand (VP3), Belgium (NSP1), Hungary (VP2, NSP2, NSP3), Italy (NSP4) and Japan (NSP5). For the strain with the G2P[4] genotype, India (VP7, VP4, VP6, NSP1, NSP4), Malawi (VP2, NSP2, NSP3), Australia (VP1), Italy (NSP5) and Bangladesh (VP3). The closest common ancestor of the strain with the genotype G4P[8] circulated in 2001-2012, depending on the gene being analyzed. For the strain with the G2P[4] genotype, the closest common ancestor dates from 2006 to 2013.
Collapse
|
87
|
Kung YH, Chi H, Liu CC, Huang YC, Huang YC, Wu FT, Huang LM. Hospital-based surveillance of severe rotavirus gastroenteritis and rotavirus strains in young Taiwanese children. J Formos Med Assoc 2020; 119:1158-1166. [PMID: 32359880 DOI: 10.1016/j.jfma.2020.03.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/17/2020] [Accepted: 03/24/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND/PURPOSE Rotavirus remains a leading cause of pediatric gastroenteritis-related hospitalization. Surveillance studies have revealed that several major rotaviral genotypes are responsible for most cases of rotavirus gastroenteritis (RVGE). This study aimed to understand the characteristics of acute gastroenteritis (AGE) caused by rotavirus in young children in Taiwan. METHODS Ten hospitals in Taiwan were subjected to prospective hospital-based AGE surveillance during 2014-2017, and children younger than 5 years old who were hospitalized due to AGE were enrolled in the study. Medical and demographic variables were recorded and analyzed, and stool specimens were collected for rotavirus identification and genotyping via real-time RT-PCR. Non-rotavirus AGE age-matched controls were enrolled. RESULTS Surveillance identified 4747 young children hospitalized with AGE during this study period. The median age of these patients was 2.0 years. Rotavirus was detected in stool samples from 518 patients (10.9%). The prevalent months of RVGE in 2014, 2015, and 2017, wherein the rotavirus positivity rates exceeded 30%. The most common serotypes were G3P[8] (303/518, 58.9%) and G1P[8] (86/518, 16.6%). The percentage of G3P[8] increased from 4.9% in 2014 to 74.3% in 2016 (P < 0.0001), whereas the percentage of G1P[8] decreased from 61.0% in 2014 to 22.5% in 2015 (P < 0.0001). Compared with G3P[8], G1P[8] was associated with a significantly higher C-reactive protein level (P < 0.05). CONCLUSION Rotavirus remains a notable pathogenic etiology of childhood AGE and the G3P[8] serotype was dominant in Taiwan. This study highlighted the importance of rotavirus surveillance to ensure protective effectiveness against the circulating strains.
Collapse
Affiliation(s)
- Yen-Hsin Kung
- Department of Pediatrics, Mackay Memorial Hospital and Mackay Children's Hospital, Taipei, Taiwan
| | - Hsin Chi
- Department of Pediatrics, Mackay Memorial Hospital and Mackay Children's Hospital, Taipei, Taiwan; Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.
| | - Ching-Chuan Liu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yhu-Chering Huang
- Department of Pediatrics, Chang Gung Children's Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yi-Chuan Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Fang-Tzy Wu
- Department of Health, Research and Diagnostic Center, Centers for Disease Control, Taiwan
| | - Li-Min Huang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
88
|
Salamanca de la Cueva I, Pahud B, Huang LM, Leonardi M, Garcia-Sicilia J, Céspedes J, Abdelnour A, Tamura T, Kuroki H, Chiu NC, Virta M, Kokko S, Horn M, Panzer F, Kim JH, Lee J, Moerman L, Debacq C, Parra J, Ugarte A, Bi D. Immunogenicity and safety of porcine circovirus-free human rotavirus vaccine in healthy infants: a phase III, randomized trial. J Infect Dis 2020; 225:jiaa210. [PMID: 32365189 PMCID: PMC9200154 DOI: 10.1093/infdis/jiaa210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/28/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Porcine circovirus type 1 (PCV-1) material was detected in the human rotavirus vaccine (HRV) in 2010. In this study (NCT02914184) we compared immunogenicity and safety of the PCV-free HRV vaccine (PCV-free HRV) with HRV. PCV-free HRV is an HRV with no detection of PCV-1 and PCV-2 according to the limit of detection of the tests used. METHODS Healthy infants 6-12 weeks of age were randomized (1:1:1:1) to receive 2 doses of 1 of the 3 lots of PCV-free HRV or HRV. The study objectives were to demonstrate lot-to-lot consistency of the PCV-free HRV and non-inferiority of PCV-free HRV as compared to HRV in terms of immunogenicity, 1-2 months post-dose 2. Reactogenicity and safety were also assessed. RESULTS Overall, 1612 infants were enrolled and 1545 completed the study. Study objectives were demonstrated since the pre-defined criteria were met. Among participants receiving PCV-free HRV and HRV, 79.27% and 81.76% seroconverted and geometric mean concentrations were 159.5 and 152.8 U/mL, respectively. The incidences of adverse events and serious adverse events were similar between the pooled PCV-free HRV and HRV groups. CONCLUSIONS The 3 PCV-free HRV lots demonstrated consistency and PCV-free HRV was non-inferior compared to HRV in terms of immunogenicity.
Collapse
Affiliation(s)
| | - Barbara Pahud
- Children’s Mercy Hospitals, Kansas City, Missouri, USA
| | - Li-Min Huang
- National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | - Arturo Abdelnour
- Instituto de Investigación en Ciencias Médicas, San José, Costa Rica
| | | | | | | | - Miia Virta
- Tampere Vaccine Research Center, Tampere University, Tampere, Finland
| | - Satu Kokko
- Tampere Vaccine Research Center, Tampere University, Tampere, Finland
| | - Michael Horn
- Pediatric Office, Schoenau am Koenigssee, Germany
| | - Falko Panzer
- Pediatric Office Dres. Panzer and Colleagues, Mannheim, Germany
| | - Jong-Hyun Kim
- St Vincent’s Hospital, Catholic University of Korea, Suwon, Republic of Korea
| | - Jin Lee
- Hanil General Hospital, Seoul, Republic of Korea
| | | | | | | | - Ana Ugarte
- Janssen Pharmaceutica companies of Johnson and Johnson, Beerse, Belgium
| | - Dan Bi
- GlaxoSmithKline, Wavre, Belgium
| | | |
Collapse
|
89
|
Fix J, Chandrashekhar K, Perez J, Bucardo F, Hudgens MG, Yuan L, Twitchell E, Azcarate-Peril MA, Vilchez S, Becker-Dreps S. Association between Gut Microbiome Composition and Rotavirus Vaccine Response among Nicaraguan Infants. Am J Trop Med Hyg 2020; 102:213-219. [PMID: 31802728 DOI: 10.4269/ajtmh.19-0355] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Rotavirus is the leading cause of childhood deaths due to diarrhea. Although existing oral rotavirus vaccines are highly efficacious in high-income countries, these vaccines have been demonstrated to have decreased efficacy in low- and middle-income countries. A possible explanation for decreased efficacy is the impact of gut microbiota on the enteric immune system's response to vaccination. We analyzed the gut microbiome of 50 children enrolled in a prospective study evaluating response to oral pentavalent rotavirus vaccination (RV5) to assess associations between relative abundance of bacterial taxa and seroconversion following vaccination. Stool samples were taken before the first RV5 dose, and microbiome composition characterized using 16S rRNA amplicon sequencing and Quantitative Insights Into Microbial Ecology software. Relative abundance of bacterial taxa between seroconverters following the first RV5 dose, those with ≥ 4-fold increase in rotavirus-specific IgA titers, and nonseroconverters were compared using the Wilcoxon-Mann-Whitney test. We identified no significant differences in microbiome composition between infants who did and did not respond to vaccination. Infants who responded to vaccination tended to have higher abundance of Proteobacteria and Eggerthella, whereas those who did not respond had higher abundance of Fusobacteria and Enterobacteriaceae; however, these differences were not statistically significant following a multiple comparison correction. This study suggests a limited impact of gut microbial taxa on response to oral rotavirus vaccination among infants; however, additional research is needed to improve our understanding of the impact of gut microbiome on vaccine response, toward a goal of improving vaccine efficacy and rotavirus prevention.
Collapse
Affiliation(s)
- Jonathan Fix
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | - Johann Perez
- Center of Infectious Diseases, Department of Microbiology and Parasitology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León (UNAN-León), León, Nicaragua
| | - Filemon Bucardo
- Center of Infectious Diseases, Department of Microbiology and Parasitology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León (UNAN-León), León, Nicaragua
| | - Michael G Hudgens
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Lijuan Yuan
- Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Erica Twitchell
- Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | | | - Samuel Vilchez
- Center of Infectious Diseases, Department of Microbiology and Parasitology, Faculty of Medical Sciences, National Autonomous University of Nicaragua, León (UNAN-León), León, Nicaragua
| | | |
Collapse
|
90
|
Boniface K, Byars SG, Cowley D, Kirkwood CD, Bines JE. Human Neonatal Rotavirus Vaccine (RV3-BB) Produces Vaccine Take Irrespective of Histo-Blood Group Antigen Status. J Infect Dis 2020; 221:1070-1078. [PMID: 31763671 PMCID: PMC7075413 DOI: 10.1093/infdis/jiz333] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/03/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND VP4 [P] genotype binding specificities of rotaviruses and differential expression of histo-blood group antigens (HBGAs) between populations may contribute to reduced efficacy against severe rotavirus disease. P[6]-based rotavirus vaccines could broaden protection in such settings, particularly in Africa, where the Lewis-negative phenotype and P[6] rotavirus strains are common. METHODS The association between HBGA status and G3P[6] rotavirus vaccine (RV3-BB) take was investigated in a phase 2A study of RV3-BB vaccine involving 46 individuals in Dunedin, New Zealand, during 2012-2014. FUT2 and FUT3 genotypes were determined from DNA extracted from stool specimens, and frequencies of positive cumulative vaccine take, defined as an RV3-BB serum immune response (either immunoglobulin A or serum neutralizing antibody) and/or stool excretion of the vaccine strain, stratified by HBGA status were determined. RESULTS RV3-BB produced positive cumulative vaccine take in 29 of 32 individuals (91%) who expressed a functional FUT2 enzyme (the secretor group), 13 of 13 (100%) who were FUT2 null (the nonsecretor group), and 1 of 1 with reduced FUT2 activity (i.e., a weak secretor); in 37 of 40 individuals (93%) who expressed a functional FUT3 enzyme (the Lewis-positive group) and 3 of 3 who were FUT3 null (the Lewis-negative group); and in 25 of 28 Lewis-positive secretors (89%), 12 of 12 Lewis-positive nonsecretors (100%), 2 of 2 Lewis-negative secretors, and 1 of 1 Lewis-negative weak secretor. CONCLUSIONS RV3-BB produced positive cumulative vaccine take irrespective of HBGA status. RV3-BB has the potential to provide an improved level of protection in settings where P[6] rotavirus disease is endemic, irrespective of the HBGA profile of the population.
Collapse
Affiliation(s)
- Karen Boniface
- Enteric Diseases Group, Murdoch Children’s Research Institute, Seattle, Washington
| | - Sean G Byars
- Melbourne School of Population and Global Health, Seattle, Washington
| | - Daniel Cowley
- Enteric Diseases Group, Murdoch Children’s Research Institute, Seattle, Washington
- Department of Pediatrics, University of Melbourne, Seattle, Washington
| | - Carl D Kirkwood
- Enteric Diseases Group, Murdoch Children’s Research Institute, Seattle, Washington
- Department of Pediatrics, University of Melbourne, Seattle, Washington
- Bill and Melinda Gates Foundation, Seattle, Washington
| | - Julie E Bines
- Enteric Diseases Group, Murdoch Children’s Research Institute, Seattle, Washington
- Department of Pediatrics, University of Melbourne, Seattle, Washington
- Department of Gastroenterology and Clinical Nutrition, Royal Children’s Hospital, Parkville, Australia
| |
Collapse
|
91
|
Harastani HH, Reslan L, Sabra A, Ali Z, Hammadi M, Ghanem S, Hajar F, Matar GM, Dbaibo GS, Zaraket H. Genetic Diversity of Human Rotavirus A Among Hospitalized Children Under-5 Years in Lebanon. Front Immunol 2020; 11:317. [PMID: 32174920 PMCID: PMC7054381 DOI: 10.3389/fimmu.2020.00317] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/07/2020] [Indexed: 12/02/2022] Open
Abstract
Human rotavirus remains a major cause of gastroenteritis worldwide despite the availability of effective vaccines. In this study, we investigated the genetic diversity of rotaviruses circulating in Lebanon. We genetically characterized the VP4 and VP7 genes encoding the outer capsid proteins of 132 rotavirus-associated gastroenteritis specimens, previously identified in hospitalized children (<5 years) from 2011 to 2013 in Lebanon. These included 43 vaccine-breakthrough specimens and the remainder were from non-vaccinated subjects. Phylogenetic analysis of VP4 and VP7 genes revealed distinct clustering compared to the vaccine strains, and several substitutions were identified in the antigenic epitopes of Lebanese specimens. No unique changes were identified in the breakthrough specimens compared to non-breakthroughs that could explain the occurrence of infection in vaccinated children. Further, we report the emergence of a rare P[8] OP354-like strain with a G9 VP7 in Lebanon, possessing high genetic variability in their VP4 compared to vaccine strains. Therefore, human rotavirus strains circulating in Lebanon and globally have accumulated numerous substitutions in their antigenic sites compared to those currently used in the licensed vaccines. The successful spread and continued genetic drift of these strains over time might undermine the effectiveness of the vaccines. The effect of such changes in the antigenic sites on vaccine efficacy remains to be assessed.
Collapse
Affiliation(s)
- Houda H Harastani
- Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Lina Reslan
- Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Ahmad Sabra
- Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Zainab Ali
- Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Moza Hammadi
- Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Soha Ghanem
- Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Farah Hajar
- Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ghassan M Matar
- Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon.,Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ghassan S Dbaibo
- Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hassan Zaraket
- Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon.,Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
92
|
Varghese L, Ezat Wan Puteh S, Schecroun N, Jahis R, Van Vlaenderen I, Standaert BA. Applying a Constrained Optimization Portfolio Model to Aid Prioritization of Public Health Interventions in Malaysia. Value Health Reg Issues 2020; 21:172-180. [PMID: 32044690 DOI: 10.1016/j.vhri.2019.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 11/17/2019] [Indexed: 11/18/2022]
Abstract
OBJECTIVES Countries have constrained healthcare budgets and must prioritize new interventions depending on health goals and time frame. This situation is relevant in the sphere of national immunization programs, for which many different vaccines are proposed, budgets are limited, and efficient choices must be made in the order of vaccine introduction. METHODS A constrained optimization (CO) model for infectious diseases was developed in which different intervention types (prophylaxis and treatment) were combined for consideration in Malaysia. Local experts defined their priority public health issues: pneumococcal disease, dengue, hepatitis B and C, rotavirus, neonatal pertussis, and cholera. Epidemiological, cost, and effectiveness data were informed from local or regionally published literature. The model aimed to maximize quality-adjusted life-year (QALY) gain through the reduction of events in each of the different diseases, under budget and intervention coverage constraints. The QALY impact of the interventions was assessed over 2 periods: lifetime and 20 years. The period of investment was limited to 15 years. RESULTS The assessment time horizon influenced the prioritization of interventions maximizing QALY gain. The incremental health gains compared with a uninformed prioritization were large for the first 8 years and declined thereafter. Rotaviral and pneumococcal vaccines were identified as key priorities irrespective of time horizon, hepatitis B immune prophylaxis and hepatitis C treatment were priorities with the lifetime horizon, and dengue vaccination replaced these with the 20-year horizon. CONCLUSIONS CO modeling is a useful tool for making economically efficient decisions within public health programs for the control of infectious diseases by helping prioritize the selection of interventions to maximize health gain under annual budget constraints.
Collapse
Affiliation(s)
| | | | - Nadia Schecroun
- Keyrus Biopharma c/o GSK, Health Economics Department, Wavre, Belgium
| | - Rohani Jahis
- Ministry of Health, Disease Control Division, Putrajaya, Malaysia
| | | | | |
Collapse
|
93
|
Hill DL, Carr EJ, Rutishauser T, Moncunill G, Campo JJ, Innocentin S, Mpina M, Nhabomba A, Tumbo A, Jairoce C, Moll HA, van Zelm MC, Dobaño C, Daubenberger C, Linterman MA. Immune system development varies according to age, location, and anemia in African children. Sci Transl Med 2020; 12:eaaw9522. [PMID: 32024802 PMCID: PMC7738197 DOI: 10.1126/scitranslmed.aaw9522] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 07/19/2019] [Accepted: 01/13/2020] [Indexed: 12/14/2022]
Abstract
Children from low- and middle-income countries, where there is a high incidence of infectious disease, have the greatest need for the protection afforded by vaccination, but vaccines often show reduced efficacy in these populations. An improved understanding of how age, infection, nutrition, and genetics influence immune ontogeny and function is key to informing vaccine design for this at-risk population. We sought to identify factors that shape immune development in children under 5 years of age from Tanzania and Mozambique by detailed immunophenotyping of longitudinal blood samples collected during the RTS,S malaria vaccine phase 3 trial. In these cohorts, the composition of the immune system is dynamically transformed during the first years of life, and this was further influenced by geographical location, with some immune cell types showing an altered rate of development in Tanzanian children compared to Dutch children enrolled in the Generation R population-based cohort study. High-titer antibody responses to the RTS,S/AS01E vaccine were associated with an activated immune profile at the time of vaccination, including an increased frequency of antibody-secreting plasmablasts and follicular helper T cells. Anemic children had lower frequencies of recent thymic emigrant T cells, isotype-switched memory B cells, and plasmablasts; modulating iron bioavailability in vitro could recapitulate the B cell defects observed in anemic children. Our findings demonstrate that the composition of the immune system in children varies according to age, geographical location, and anemia status.
Collapse
Affiliation(s)
- Danika L Hill
- Lymphocyte Signalling and Development, Babraham Institute, Cambridge CB22 3AT, UK.
- Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Victoria 3004, Australia
| | - Edward J Carr
- Lymphocyte Signalling and Development, Babraham Institute, Cambridge CB22 3AT, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Tobias Rutishauser
- Swiss Tropical and Public Health Institute, Basel 4051, Switzerland
- University of Basel, Basel 4001, Switzerland
| | - Gemma Moncunill
- ISGlobal, Barcelona Centre for International Health Research, Hospital Clínic-Universitat de Barcelona, Catalonia 08036, Spain
| | - Joseph J Campo
- ISGlobal, Barcelona Centre for International Health Research, Hospital Clínic-Universitat de Barcelona, Catalonia 08036, Spain
| | - Silvia Innocentin
- Lymphocyte Signalling and Development, Babraham Institute, Cambridge CB22 3AT, UK
| | - Maxmillian Mpina
- Swiss Tropical and Public Health Institute, Basel 4051, Switzerland
- University of Basel, Basel 4001, Switzerland
- Ifakara Health Institute, Bagamoyo, Tanzania
| | - Augusto Nhabomba
- Centro de Investigação em Saúde de Manhiça, Maputo, CP 1929, Mozambique
| | - Anneth Tumbo
- Swiss Tropical and Public Health Institute, Basel 4051, Switzerland
- University of Basel, Basel 4001, Switzerland
- Ifakara Health Institute, Bagamoyo, Tanzania
| | - Chenjerai Jairoce
- Centro de Investigação em Saúde de Manhiça, Maputo, CP 1929, Mozambique
| | - Henriëtte A Moll
- Department of Pediatrics, Sophia Children's Hospital, Erasmus MC, University Medical Center, Rotterdam 3015 GD, Netherlands
| | - Menno C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Victoria 3004, Australia
| | - Carlota Dobaño
- ISGlobal, Barcelona Centre for International Health Research, Hospital Clínic-Universitat de Barcelona, Catalonia 08036, Spain
- Centro de Investigação em Saúde de Manhiça, Maputo, CP 1929, Mozambique
| | - Claudia Daubenberger
- Swiss Tropical and Public Health Institute, Basel 4051, Switzerland.
- University of Basel, Basel 4001, Switzerland
| | - Michelle A Linterman
- Lymphocyte Signalling and Development, Babraham Institute, Cambridge CB22 3AT, UK.
| |
Collapse
|
94
|
Rotavirus vaccination in the neonatal intensive care units: where are we? A rapid review of recent evidence. Curr Opin Pediatr 2020; 32:167-191. [PMID: 31851055 DOI: 10.1097/mop.0000000000000869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Rotavirus is a leading cause of viral acute gastroenteritis in infants. Neonates hospitalized in neonatal intensive care units (NICUs) are at risk of rotavirus infections with severe outcomes. The administration of rotavirus vaccines is only recommended, in the United States and Canada, upon discharge from the NICU despite rotavirus vaccines being proven well tolerated and effective in these populations, because of risks of live-attenuated vaccine administration in immunocompromised patients and theoretical risks of rotavirus vaccine strains shedding and transmission.We aimed to summarize recent evidence regarding rotavirus vaccine administration in the NICU setting and safety of rotavirus vaccines in preterm infants. METHODS We conducted a rapid review of the literature from the past 10 years, searching Medline and Embase, including all study types except reviews, reporting on rotavirus vaccines 1 and 5; NICU setting; shedding or transmission; safety in preterm. One reviewer performed data extraction and quality assessment. RECENT FINDINGS Thirty-one articles were analyzed. Vaccine-derived virus shedding following rotavirus vaccines existed for nearly all infants, mostly during the first week after dose 1, but with rare transmission only described in the household setting. No case of transmission in the NICU was reported. Adverse events were mild to moderate, occurring in 10-60% of vaccinated infants. Extreme premature infants or those with underlying gastrointestinal failure requiring surgery presented with more severe adverse events. SUMMARY Recommendations regarding rotavirus vaccine administration in the NICU should be reassessed in light of the relative safety and absence of transmission of rotavirus vaccine strains in the NICU.
Collapse
|
95
|
Jia S, Li J, Liu Y, Zhu F. Precision immunization: a new trend in human vaccination. Hum Vaccin Immunother 2020; 16:513-522. [PMID: 31545124 DOI: 10.1080/21645515.2019.1670123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Vaccination has been one of the major revolutions in the history of human health. Vaccination programs have targeted entire populations such as infants or elderly subjects as a matter of being efficient with time and resources. These general populations are heterogeneous in terms of factors such as ethnicity, health status, and socio-economics. Thus, there have been variations in the safety and effectiveness profiles of certain vaccinations according to current population-wide strategies. As the concept of precision medicine has been raised in recent years, many researchers have suggested that vaccines could be administered more precisely in terms of particular target populations, vaccine formulations, regimens, and dosage levels. This review addresses the concept and framework of precision immunization, summarizes recent and representative clinical trials of among specific populations, mentions important factors to be addressed in customizing vaccinations, and provides suggestions on the establishment of precision immunization with the goal of maximizing the effectiveness of vaccines in general.
Collapse
Affiliation(s)
- Siyue Jia
- Vaccine Clinical Evaluation Department, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, PR China
| | - Jingxin Li
- Vaccine Clinical Evaluation Department, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, PR China
| | - Yuanbao Liu
- Expanded Program on Immunization Department, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, PR China.,NHC Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, PR China
| | - Fengcai Zhu
- Vaccine Clinical Evaluation Department, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, PR China.,NHC Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, PR China
| |
Collapse
|
96
|
Jere KC, Bar-Zeev N, Chande A, Bennett A, Pollock L, Sanchez-Lopez PF, Nakagomi O, Tate JE, Parashar UD, Heyderman RS, French N, Iturriza-Gomara M, Cunliffe NA. Vaccine Effectiveness against DS-1-Like Rotavirus Strains in Infants with Acute Gastroenteritis, Malawi, 2013-2015. Emerg Infect Dis 2020; 25:1734-1737. [PMID: 31441761 PMCID: PMC6711242 DOI: 10.3201/eid2509.190258] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Atypical DS-1-like G1P[8] rotaviruses emerged in 2013 in Malawi after rotavirus vaccine introduction. Vaccine effectiveness among infants hospitalized with acute DS-1-like G1P[8] rotavirus gastroenteritis was 85.6% (95% CI 34.4%-96.8%). These findings suggest that vaccine provides protection against these strains despite their emergence coinciding with vaccine introduction.
Collapse
|
97
|
Sen A, Ding S, Greenberg HB. The Role of Innate Immunity in Regulating Rotavirus Replication, Pathogenesis, and Host Range Restriction and the Implications for Live Rotaviral Vaccine Development. MUCOSAL VACCINES 2020. [PMCID: PMC7148637 DOI: 10.1016/b978-0-12-811924-2.00041-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Rotaviruses (RVs) are important causative agents of viral gastroenteritis in the young of most mammalian species studied, including humans, in which they are the most important cause of severe gastroenteritis worldwide despite the availability of several safe and effective vaccines. Replication of RVs is restricted in a host species-specific manner, and this barrier is determined predominantly by the host interferon (IFN) signaling and the ability of different RV strains to successfully negate IFN activation and amplification pathways. In addition, viral attachment to the target intestinal epithelial cells also regulates host range restriction. Several studies have focused on the role of the innate immune response in regulating RV replication and pathogenesis. The knowledge accrued from these efforts is likely to result in rational attenuation of RV vaccines to closely match circulating (and host species-matched) virus strains. In this chapter, we review prevalent models of RV interactions with innate immune factors, viral strategies employed to regulate their function, and the implications of these findings for improved RV vaccine development.
Collapse
|
98
|
Motamedi-Rad M, Farahmand M, Arashkia A, Jalilvand S, Shoja Z. VP7 and VP4 genotypes of rotaviruses cocirculating in Iran, 2015 to 2017: Comparison with cogent sequences of Rotarix and RotaTeq vaccine strains before their use for universal mass vaccination. J Med Virol 2019; 92:1110-1123. [PMID: 31774174 DOI: 10.1002/jmv.25642] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 11/23/2019] [Indexed: 12/17/2022]
Abstract
The present study was conducted to analyze the genotypic diversity of circulating species A rotavirus (RVA) strains in Iran and also to investigate comparative analysis between the genotypes of VP4 and VP7 of cocirculating RVA and vaccine strains before the vaccine is introduced in the national immunization program. The G3-lineage I was found in this study as the most common G genotype which was followed by G9-lineage III, G1-lineages I, II, G12-lineage III, G2-lineage IV, and G4-lineage I. Also, P[8]-lineages III, IV was found as the predominant P genotype which was followed by P[4]-lineage V, and P[6]-lineage I. Overally, G3P[8] was determined as the most common combination. Moreover, the analysis of the VP7 antigenic epitopes showed that several amino acid differences existed between circulating Iranian and the vaccine strains. The comparison of genotype G1 of Iranian and vaccine strains (RotaTeq and Rotarix), and genotypes G2, G3, and G4 of Iranian and RotaTeq vaccine strains revealed three to five amino acids differences on the VP7 antigenic epitopes. Furthermore, analyzing of the VP8* epitopes of Iranian P[8] strains indicated that they contained up to 11 and 14 amino acid differences with Rotarix and RotaTeq, respectively. Based on different patterns of amino acid substitutions in circulating and vaccine strains, the emergence of antibody escaping mutants and potentially the decrease of immune protection might ensue in vaccinated children. However, considering the broad cross-protective activity of RVA vaccines, their efficacy should be monitored after the introduction in Iran.
Collapse
Affiliation(s)
| | - Mohammad Farahmand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Arashkia
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Somayeh Jalilvand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Zabihollah Shoja
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
99
|
Antirotavirus IgA seroconversion rates in children who receive concomitant oral poliovirus vaccine: A secondary, pooled analysis of Phase II and III trial data from 33 countries. PLoS Med 2019; 16:e1003005. [PMID: 31887139 PMCID: PMC6936798 DOI: 10.1371/journal.pmed.1003005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/22/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Despite the success of rotavirus vaccines over the last decade, rotavirus remains a leading cause of severe diarrheal disease among young children. Further progress in reducing the burden of disease is inhibited, in part, by vaccine underperformance in certain settings. Early trials suggested that oral poliovirus vaccine (OPV), when administered concomitantly with rotavirus vaccine, reduces rotavirus seroconversion rates after the first rotavirus dose with modest or nonsignificant interference after completion of the full rotavirus vaccine course. Our study aimed to identify a range of individual-level characteristics, including concomitant receipt of OPV, that affect rotavirus vaccine immunogenicity in high- and low-child-mortality settings, controlling for individual- and country-level factors. Our central hypothesis was that OPV administered concomitantly with rotavirus vaccine reduced rotavirus vaccine immunogenicity. METHODS AND FINDINGS Pooled, individual-level data from GlaxoSmithKline's Phase II and III clinical trials of the monovalent rotavirus vaccine (RV1), Rotarix, were analyzed, including 7,280 vaccinated infants (5-17 weeks of age at first vaccine dose) from 22 trials and 33 countries/territories (5 countries/territories with high, 13 with moderately low, and 15 with very low child mortality). Two standard markers for immune response were examined including antirotavirus immunoglobulin A (IgA) seroconversion (defined as the appearance of serum antirotavirus IgA antibodies in subjects initially seronegative) and serum antirotavirus IgA titer, both collected approximately 4-12 weeks after administration of the last rotavirus vaccine dose. Mixed-effect logistic regression and mixed-effect linear regression of log-transformed data were used to identify individual- and country-level predictors of seroconversion (dichotomous) and antibody titer (continuous), respectively. Infants in high-child-mortality settings had lower odds of seroconverting compared with infants in low-child-mortality settings (odds ratio [OR] = 0.48, 95% confidence interval [CI] 0.43-0.53, p < 0.001). Similarly, among those who seroconverted, infants in high-child-mortality settings had lower IgA titers compared with infants in low-child-mortality settings (mean difference [β] = 0.83, 95% CI 0.77-0.90, p < 0.001). Infants who received OPV concomitantly with both their first and their second doses of rotavirus vaccine had 0.63 times the odds of seroconverting (OR = 0.63, 95% CI 0.47-0.84, p = 0.002) compared with infants who received OPV but not concomitantly with either dose. In contrast, among infants who seroconverted, OPV concomitantly administered with both the first and second rotavirus vaccine doses was found to be positively associated with antirotavirus IgA titer (β = 1.28, 95% CI 1.07-1.53, p = 0.009). Our findings may have some limitations in terms of generalizability to routine use of rotavirus vaccine because the analysis was limited to healthy infants receiving RV1 in clinical trial settings. CONCLUSIONS Our findings suggest that OPV given concomitantly with RV1 was a substantial contributor to reduced antirotavirus IgA seroconversion, and this interference was apparent after the second vaccine dose of RV1, as with the original clinical trials that our reanalysis is based on. However, our findings do suggest that the forthcoming withdrawal of OPV from the infant immunization schedule globally has the potential to improve RV1 performance.
Collapse
|
100
|
Parenterally Administered P24-VP8* Nanoparticle Vaccine Conferred Strong Protection against Rotavirus Diarrhea and Virus Shedding in Gnotobiotic Pigs. Vaccines (Basel) 2019; 7:vaccines7040177. [PMID: 31698824 PMCID: PMC6963946 DOI: 10.3390/vaccines7040177] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 12/23/2022] Open
Abstract
Current live rotavirus vaccines are costly with increased risk of intussusception due to vaccine replication in the gut of vaccinated children. New vaccines with improved safety and cost-effectiveness are needed. In this study, we assessed the immunogenicity and protective efficacy of a novel P24-VP8* nanoparticle vaccine using the gnotobiotic (Gn) pig model of human rotavirus infection and disease. Three doses of P24-VP8* (200 μg/dose) intramuscular vaccine with Al(OH)3 adjuvant (600 μg) conferred significant protection against infection and diarrhea after challenge with virulent Wa strain rotavirus. This was indicated by the significant reduction in the mean duration of diarrhea, virus shedding in feces, and significantly lower fecal cumulative consistency scores in post-challenge day (PCD) 1-7 among vaccinated pigs compared to the mock immunized controls. The P24-VP8* vaccine was highly immunogenic in Gn pigs. It induced strong VP8*-specific serum IgG and Wa-specific virus-neutralizing antibody responses from post-inoculation day 21 to PCD 7, but did not induce serum or intestinal IgA antibody responses or a strong effector T cell response, which are consistent with the immunization route, the adjuvant used, and the nature of the non-replicating vaccine. The findings are highly translatable and thus will facilitate clinical trials of the P24-VP8* nanoparticle vaccine.
Collapse
|