Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Gen Virol ; 99(4): 457-463, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29517476

RESUMO

Rotavirus B (RVB) has been associated with enteric disease in many animal species. An RVB strain was identified in pooled intestinal samples from Alpine caprine kids (between 2 and 3 days of age) experiencing high (>90 %) morbidity, and the complete caprine RVB genome was characterized. Histology revealed villus atrophy, the samples tested positive for RVB by real-time RT-PCR and metagenomic next-generation sequencing identified only RVB and orf virus. In the VP4 gene segment, the caprine RVB strain had a higher percentage nucleotide identity to the Indian bovine RVB strains than to the Japanese bovine RVB strains, but the VP7, VP6, VP2, NSP1, NSP2 and NSP5 gene segments of the American caprine RVB strain were genetically related to the Japanese bovine RVB strains. The results indicate a lack of RVB sequences to understand reassortment or the evolutionary relationship of RVB strains from cattle and goats.


Assuntos
Doenças dos Bovinos/virologia , Enterite/veterinária , Genoma Viral , Doenças das Cabras/virologia , Infecções por Rotavirus/veterinária , Rotavirus/genética , Animais , Animais Recém-Nascidos/virologia , Bovinos , Doenças dos Bovinos/transmissão , Enterite/virologia , Genótipo , Doenças das Cabras/transmissão , Cabras , Filogenia , Rotavirus/química , Rotavirus/classificação , Rotavirus/isolamento & purificação , Infecções por Rotavirus/transmissão , Infecções por Rotavirus/virologia , Proteínas Virais/química , Proteínas Virais/genética
2.
J Gen Virol ; 98(12): 2931-2936, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29168675

RESUMO

Rotavirus G (RVG) strains have been detected in a variety of avian species, but RVG genomes have been published from only a single pigeon and two chicken strains. Two turkey RVG strains were identified and characterized, one in a hatchery with no reported health issues and the other in a hatchery with high embryo/poult mortality. The two turkey RVG strains shared only an 85.3 % nucleotide sequence identity in the VP7 gene while the other genes possessed high nucleotide identity among them (96.3-99.9 %). Low nucleotide percentage identities (31.6-87.3 %) occurred among the pigeon and chicken RVG strains. Interestingly, potential recombination events were detected between our RVG strains and a human RVB strain, in the VP6 and NSP3 segments. The epidemiology of RVG in avian flocks and the pathogenicity of the two different RVG strains should be further investigated to understand the ecology and impact of RVG in commercial poultry flocks.


Assuntos
Genoma Viral , Filogenia , Doenças das Aves Domésticas/epidemiologia , Recombinação Genética , Infecções por Rotavirus/veterinária , Rotavirus/genética , Animais , Antígenos Virais/genética , Antígenos Virais/metabolismo , Sequência de Bases , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Galinhas/virologia , China/epidemiologia , Columbidae/virologia , Embrião não Mamífero , Humanos , Doenças das Aves Domésticas/transmissão , Doenças das Aves Domésticas/virologia , Rotavirus/classificação , Infecções por Rotavirus/epidemiologia , Infecções por Rotavirus/transmissão , Infecções por Rotavirus/virologia , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Perus/virologia , Estados Unidos/epidemiologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
3.
J Infect Dis ; 214(7): 1039-49, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27471320

RESUMO

BACKGROUND: P[6] rotaviruses have been circulating with a high prevalence in African and, to a more limited extent, Asian countries, but they have not been highly prevalent in other parts of the world. METHODS: To investigate the genomic relationship between African and Asian human P[6] rotaviruses and P[4] and P[8] rotaviruses circulating worldwide, we sequenced 39 P[6] strains, collected in Ghana, Mali, Kenya and Bangladesh, providing the largest data set of P[6] rotavirus genomes isolated in low-income countries or anywhere else in the world that has been published thus far. RESULTS: Overall, the data indicate that the genetic backbone of human P[6] strains from the low-income countries are similar to those of P[4] or P[8] strains circulating worldwide. CONCLUSIONS: The observation that gene segment 4 is the main differentiator between human P[6] and non-P[6] strains suggests that the VP4 spike protein is most likely one of the main reasons preventing the rapid spread of P[6] strains to the rest of the world despite multiple introductions. These observations reinforce previous findings about the receptor specificity of P[6] rotavirus strains.


Assuntos
Variação Genética , Genótipo , Infecções por Rotavirus/virologia , Rotavirus/classificação , Rotavirus/genética , África Subsaariana/epidemiologia , Sudeste Asiático/epidemiologia , Análise por Conglomerados , Humanos , Epidemiologia Molecular , Filogeografia , RNA Viral/genética , Rotavirus/isolamento & purificação , Infecções por Rotavirus/epidemiologia , Análise de Sequência de DNA , Homologia de Sequência
4.
Emerg Infect Dis ; 20(7): 1195-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24960190

RESUMO

We investigated the presence in US pigs of rotavirus H (RVH), identified in pigs in Japan and Brazil. From 204 samples collected during 2006-2009, we identified RVH in 15% of fecal samples from 10 US states, suggesting that RVH has circulated in the United States since 2002, but probably longer.


Assuntos
Infecções por Rotavirus/virologia , Rotavirus/genética , Doenças dos Suínos/virologia , Suínos/virologia , Animais , Fezes/virologia , Japão , Filogenia , Análise de Sequência de DNA/métodos , Estados Unidos
5.
J Gen Virol ; 93(Pt 4): 866-875, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22190012

RESUMO

In this study, the complete genome sequences of seven equine group A rotavirus (RVA) strains (RVA/Horse-tc/GBR/L338/1991/G13P[18], RVA/Horse-wt/IRL/03V04954/2003/G3P[12] and RVA/Horse-wt/IRL/04V2024/2004/G14P[12] from Europe; RVA/Horse-wt/ARG/E30/1993/G3P[12], RVA/Horse-wt/ARG/E403/2006/G14P[12] and RVA/Horse-wt/ARG/E4040/2008/G14P[12] from Argentina; and RVA/Horse-wt/ZAF/EqRV-SA1/2006/G14P[12] from South Africa) were determined. Multiple novel genotypes were identified and genotype numbers were assigned by the Rotavirus Classification Working Group: R9 (VP1), C9 (VP2), N9 (NSP2), T12 (NSP3), E14 (NSP4), and H7 and H11 (NSP5). The genotype constellation of L338 was unique: G13-P[18]-I6-R9-C9-M6-A6-N9-T12-E14-H11. The six remaining equine RVA strains showed a largely conserved genotype constellation: G3/G14-P[12]-I2/I6-R2-C2-M3-A10-N2-T3-E2/E12-H7, which is highly divergent from other known non-equine RVA genotype constellations. Phylogenetic analyses revealed that the sequences of these equine RVA strains are related distantly to non-equine RVA strains, and that at least three lineages exist within equine RVA strains. A small number of reassortment events were observed. Interestingly, the three RVA strains from Argentina possessed the E12 genotype, whereas the three RVA strains from Ireland and South Africa possessed the E2 genotype. The unusual E12 genotype has until now only been described in Argentina among RVA strains collected from guanaco, cattle and horses, suggesting geographical isolation of this NSP4 genotype. This conserved genetic configuration of equine RVA strains could be useful for future vaccine development or improvement of currently used equine RVA vaccines.


Assuntos
Sequência Conservada/genética , Gastroenterite/veterinária , Genoma Viral/genética , Doenças dos Cavalos/virologia , Infecções por Rotavirus/veterinária , Rotavirus/genética , Animais , Sequência de Bases , Gastroenterite/virologia , Genes Virais/genética , Genótipo , Cavalos/virologia , Dados de Sequência Molecular , Filogenia , Infecções por Rotavirus/virologia
6.
J Clin Microbiol ; 50(3): 966-76, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22189107

RESUMO

Two live-attenuated rotavirus group A (RVA) vaccines, Rotarix (G1P[8]) and RotaTeq (G1-G4, P[8]), have been successfully introduced in many countries worldwide, including Belgium. The parental RVA strains used to generate the vaccines were isolated more than 20 years ago in France (G4 parental strain in RotaTeq) and the United States (all other parental strains). At present, little is known about the relationship between currently circulating human RVAs and the vaccine strains. In this study, we determined sequences for the VP7 and VP4 outer capsid proteins of representative G1P[8], G2P[4], G3P[8], G4P[8], G9P[8], and G12P[8] RVAs circulating in Belgium during 2007 to 2009. The analyses showed that multiple amino acid differences existed between the VP7 and VP4 antigenic epitopes of the vaccine viruses and the Belgian isolates, regardless of their G and P genotypes. However, the highest variability was observed among the circulating G1P[8] RVA strains and the G1 and P[8] components of both RVA vaccines. In particular, RVA strains of the P[8] lineage 4 (OP354-like) showed a significant number of amino acid differences with the P[8] VP4 of both vaccines. In addition, the circulating Belgian G3 RVA strains were found to possibly possess an extra N-linked glycosylation site compared to the G3 RVA vaccine strain of RotaTeq. These results indicate that the antigenic epitopes of RVA strains contained in the vaccines differ substantially from those of the currently circulating RVA strains in Belgium. Over time, these differences might result in selection for strains that escape the RVA neutralizing-antibody pressure induced by vaccines.


Assuntos
Antígenos Virais/imunologia , Proteínas do Capsídeo/imunologia , Epitopos/imunologia , Infecções por Rotavirus/epidemiologia , Vacinas contra Rotavirus/imunologia , Rotavirus/imunologia , Substituição de Aminoácidos , Antígenos Virais/genética , Bélgica/epidemiologia , Proteínas do Capsídeo/genética , Estudos Epidemiológicos , Epitopos/genética , Humanos , Epidemiologia Molecular , Mutação de Sentido Incorreto , RNA Viral/genética , Rotavirus/genética , Rotavirus/isolamento & purificação , Infecções por Rotavirus/virologia , Análise de Sequência de DNA , Vacinas Atenuadas/imunologia
7.
Arch Virol ; 157(6): 1177-82, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22430951

RESUMO

Indirect immunofluorescence techniques targeting the rotavirus (RV) protein VP6 are used to differentiate RV species. The ICTV recognizes RV species A to E and two tentative species, F and G. A potential new RV species, ADRV-N, has been described. Phylogenetic trees and pairwise identity frequency graphs were constructed with more than 400 available VP6 sequences and seven newly determined VP6 sequences of RVD strains. All RV species were separated into distinct phylogenetic clusters. An amino acid sequence cutoff value of 53% firmly permitted differentiation of RV species, and ADRV-N was tentatively assigned to a novel RV species H (RVH).


Assuntos
Antígenos Virais/genética , Proteínas do Capsídeo/genética , Doenças das Aves Domésticas/virologia , Infecções por Rotavirus/veterinária , Rotavirus/classificação , Rotavirus/isolamento & purificação , Sequência de Aminoácidos , Animais , Antígenos Virais/química , Proteínas do Capsídeo/química , Galinhas , Dados de Sequência Molecular , Filogenia , Rotavirus/química , Rotavirus/genética , Infecções por Rotavirus/virologia , Homologia de Sequência de Aminoácidos
8.
Viruses ; 14(11)2022 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-36423115

RESUMO

Rotavirus A (RVA) possesses a genome of 11 double-stranded (ds) RNA segments, and each segment encodes one protein, with the exception of segment 11. NSP4 is a non-structural multifunctional protein encoded by segment 10 that defines the E-genotype. From the 31 E-genotypes described, genotype E12 has been described in Argentina, Uruguay, Paraguay, and Brazil in RVA strains infecting different animal species and humans. In this work, we studied the evolutionary relationships of RVA strains carrying the E12 genotype in South America using phylogenetic and phylodynamic approaches. We found that the E12 genotype has a South American origin, with a guanaco (Lama guanicoe) strain as natural host. Interestingly, all the other reported RVA strains carrying the E12 genotype in equine, bovine, caprine, and human strains are related to RVA strains of camelid origin. The evolutionary path and genetic footprint of the E12 genotype were reconstructed starting with the introduction of non-native livestock species into the American continent with the Spanish conquest in the 16th century. The imported animal species were in close contact with South American camelids, and the offspring were exposed to the native RVA strains brought from Europe and the new RVA circulating in guanacos, resulting in the emergence of new RVA strains in the current lineages' strongly species-specific adaption. In conclusion, we proposed the NSP4 E12 genotype as a genetic geographic marker in the RVA strains circulating in different animal species in South America.


Assuntos
Camelídeos Americanos , Infecções por Rotavirus , Rotavirus , Animais , Bovinos , Cavalos , Humanos , Rotavirus/genética , Filogenia , Cabras , Genótipo , Brasil
9.
Pathogens ; 11(4)2022 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-35456099

RESUMO

Sicily was the first Italian region to introduce rotavirus (RV) vaccination with the monovalent G1P[8] vaccine Rotarix® in May 2012. In this study, the seasonal distribution and molecular characterization of RV strains detected over 19 years were compared to understand the effect of Rotarix® on the evolutionary dynamics of human RVs. A total of 7846 stool samples collected from children < 5 years of age, hospitalized with acute gastroenteritis, were tested for RV detection and genotyping. Since 2013, vaccine coverage has progressively increased, while the RV prevalence decreased from 36.1% to 13.3% with a loss of seasonality. The local distribution of RV genotypes changed over the time possibly due to vaccine introduction, with a drastic reduction in G1P[8] strains replaced by common and novel emerging RV strains, such as equine-like G3P[8] in the 2018−2019 season. Comparison of VP7 and VP4 amino acid (aa) sequences with the cognate genes of Rotarix® and RotaTeq® vaccine strains showed specific aa changes in the antigenic epitopes of VP7 and of the VP8* portion of VP4 of the Italian RV strains. Molecular epidemiological surveillance data are required to monitor the emergence of novel RV strains and ascertain if these strains may affect the efficacy of RV vaccines.

10.
Lancet ; 376(9741): 606-14, 2010 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-20692030

RESUMO

BACKGROUND: Rotavirus gastroenteritis causes many deaths in infants in sub-Saharan Africa. Because rotavirus vaccines have proven effective in developed countries but had not been tested in developing countries, we assessed efficacy of a pentavalent rotavirus vaccine against severe disease in Ghana, Kenya, and Mali between April, 2007, and March, 2009. METHODS: In our multicentre, double-blind, placebo-controlled trial, undertaken in rural areas of Ghana and Kenya and an urban area of Mali, we randomly assigned infants aged 4-12 weeks without symptoms of gastrointestinal disorders in a 1:1 ratio to receive three oral doses of pentavalent rotavirus vaccine 2 mL or placebo at around 6 weeks, 10 weeks, and 14 weeks of age. Infants with HIV infection were not excluded. Randomisation was done by computer-generated randomisation sequence in blocks of six. We obtained data for gastrointestinal symptoms from parents on presentation to health-care facilities and clinical data were obtained prospectively by clinicians. The primary endpoint was severe rotavirus gastroenteritis (Vesikari score >or=11), detected by enzyme immunoassay, arising 14 days or more after the third dose of placebo or vaccine to end of study (March 31, 2009; around 21 months of age). Analysis was per protocol; infants who received scheduled doses of vaccine or placebo without intervening laboratory-confirmed naturally occurring rotavirus disease earlier than 14 days after the third dose and had complete clinical and laboratory results were included in the analysis. This study is registered with ClinicalTrials.gov, number NCT00362648. FINDINGS: 5468 infants were randomly assigned to receive pentavalent rotavirus vaccine (n=2733) or placebo (n=2735). 2357 infants assigned to vaccine and 2348 assigned to placebo were included in the per-protocol analysis. 79 cases of severe rotavirus gastroenteritis were reported in 2610.6 person-years in the vaccine group, compared with 129 cases in 2585.9 person-years in the placebo group, resulting in a vaccine efficacy against severe rotavirus gastroenteritis of 39.3% (95% CI 19.1-54.7, p=0.0003 for efficacy >0%). Median follow-up in both groups was 527 days starting 14 days after the third dose of vaccine or placebo was given. 42 (1.5%) of 2723 infants assigned to receive vaccine and 45 (1.7%) of 2724 infants assigned to receive placebo had a serious adverse event within 14 days of any dose. The most frequent serious adverse event was gastroenteritis (vaccine 17 [0.6%]; placebo 17 [0.6%]). INTERPRETATION: Pentavalent rotavirus vaccine is effective against severe rotavirus gastroenteritis in the first 2 years of life in African countries with high mortality in infants younger than 5 years. We support WHO's recommendation for adoption of rotavirus vaccine into national expanded programmes on immunisation in Africa. FUNDING: PATH (GAVI Alliance grant) and Merck.


Assuntos
Países em Desenvolvimento , Gastroenterite/prevenção & controle , Infecções por Rotavirus/prevenção & controle , Vacinas contra Rotavirus/administração & dosagem , Administração Oral , África Subsaariana , Anticorpos Antivirais/sangue , Método Duplo-Cego , Gastroenterite/virologia , Humanos , Esquemas de Imunização , Imunoglobulina A/sangue , Lactente , Rotavirus/imunologia , Infecções por Rotavirus/imunologia , Vacinas contra Rotavirus/imunologia , Índice de Gravidade de Doença , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia
11.
Lancet ; 376(9741): 615-23, 2010 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-20692031

RESUMO

BACKGROUND: Rotavirus vaccine has proved effective for prevention of severe rotavirus gastroenteritis in infants in developed countries, but no efficacy studies have been done in developing countries in Asia. We assessed the clinical efficacy of live oral pentavalent rotavirus vaccine for prevention of severe rotavirus gastroenteritis in infants in Bangladesh and Vietnam. METHODS: In this multicentre, double-blind, placebo-controlled trial, undertaken in rural Matlab, Bangladesh, and urban and periurban Nha Trang, Vietnam, infants aged 4-12 weeks without symptoms of gastrointestinal disorders were randomly assigned (1:1) to receive three oral doses of pentavalent rotavirus vaccine 2 mL or placebo at around 6 weeks, 10 weeks, and 14 weeks of age, in conjunction with routine infant vaccines including oral poliovirus vaccine. Randomisation was done by computer-generated randomisation sequence in blocks of six. Episodes of gastroenteritis in infants who presented to study medical facilities were reported by clinical staff and from parent recollection. The primary endpoint was severe rotavirus gastroenteritis (Vesikari score >or=11) arising 14 days or more after the third dose of placebo or vaccine to end of study (March 31, 2009; around 21 months of age). Analysis was per protocol; infants who received scheduled doses of vaccine or placebo without intervening laboratory-confirmed naturally occurring rotavirus disease earlier than 14 days after the third dose and had complete clinical and laboratory results were included in the analysis. This study is registered with ClinicalTrials.gov, number NCT00362648. FINDINGS: 2036 infants were randomly assigned to receive pentavalent rotavirus vaccine (n=1018) or placebo (n=1018). 991 infants assigned to pentavalent rotavirus vaccine and 978 assigned to placebo were included in the per-protocol analysis. Median follow up from 14 days after the third dose of placebo or vaccine until final disposition was 498 days (IQR 480-575). 38 cases of severe rotavirus gastroenteritis (Vesikari score >or=11) were reported during more than 1197 person-years of follow up in the vaccine group, compared with 71 cases in more than 1156 person years in the placebo group, resulting in a vaccine efficacy of 48.3% (95% CI 22.3-66.1) against severe disease (p=0.0005 for efficacy >0%) during nearly 2 years of follow-up. 25 (2.5%) of 1017 infants assigned to receive vaccine and 20 (2.0%) of 1018 assigned to receive placebo had a serious adverse event within 14 days of any dose. The most frequent serious adverse event was pneumonia (vaccine 12 [1.2%]; placebo 15 [1.5%]). INTERPRETATION: In infants in developing countries in Asia, pentavalent rotavirus vaccine is safe and efficacious against severe rotavirus gastroenteritis, and our results support expanded WHO recommendations to promote its global use. FUNDING: PATH (GAVI Alliance grant) and Merck.


Assuntos
Países em Desenvolvimento , Gastroenterite/prevenção & controle , Infecções por Rotavirus/prevenção & controle , Vacinas contra Rotavirus/administração & dosagem , Administração Oral , Anticorpos Antivirais/sangue , Bangladesh , Método Duplo-Cego , Feminino , Gastroenterite/virologia , Humanos , Esquemas de Imunização , Imunoglobulina A/sangue , Lactente , Masculino , Infecções por Rotavirus/imunologia , Vacinas contra Rotavirus/imunologia , Índice de Gravidade de Doença , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Vietnã
12.
Arch Virol ; 156(8): 1397-413, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21597953

RESUMO

In April 2008, a nucleotide-sequence-based, complete genome classification system was developed for group A rotaviruses (RVs). This system assigns a specific genotype to each of the 11 genome segments of a particular RV strain according to established nucleotide percent cutoff values. Using this approach, the genome of individual RV strains are given the complete descriptor of Gx-P[x]-Ix-Rx-Cx-Mx-Ax-Nx-Tx-Ex-Hx. The Rotavirus Classification Working Group (RCWG) was formed by scientists in the field to maintain, evaluate and develop the RV genotype classification system, in particular to aid in the designation of new genotypes. Since its conception, the group has ratified 51 new genotypes: as of April 2011, new genotypes for VP7 (G20-G27), VP4 (P[28]-P[35]), VP6 (I12-I16), VP1 (R5-R9), VP2 (C6-C9), VP3 (M7-M8), NSP1 (A15-A16), NSP2 (N6-N9), NSP3 (T8-T12), NSP4 (E12-E14) and NSP5/6 (H7-H11) have been defined for RV strains recovered from humans, cows, pigs, horses, mice, South American camelids (guanaco), chickens, turkeys, pheasants, bats and a sugar glider. With increasing numbers of complete RV genome sequences becoming available, a standardized RV strain nomenclature system is needed, and the RCWG proposes that individual RV strains are named as follows: RV group/species of origin/country of identification/common name/year of identification/G- and P-type. In collaboration with the National Center for Biotechnology Information (NCBI), the RCWG is also working on developing a RV-specific resource for the deposition of nucleotide sequences. This resource will provide useful information regarding RV strains, including, but not limited to, the individual gene genotypes and epidemiological and clinical information. Together, the proposed nomenclature system and the NCBI RV resource will offer highly useful tools for investigators to search for, retrieve, and analyze the ever-growing volume of RV genomic data.


Assuntos
Rotavirus/classificação , Rotavirus/genética , Terminologia como Assunto , Animais , Genoma Viral , Genótipo , Humanos , Especificidade da Espécie
13.
Hum Vaccin ; 7(5): 506-10, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21422820

RESUMO

RotaTeq is an oral pentavalent rotavirus vaccine (RV5) that has shown high and consistent efficacy in preventing rotavirus gastroenteritis (RGE) in randomized clinical trials conducted mostly in industrialized countries. We projected the effectiveness of RV5 against RGE-related hospitalizations and deaths in 6 Asian countries by using a simple mathematical model. Model inputs included rotavirus surveillance data collected 2006-2007 in China, 2001-2002 in Hong Kong, 2005-2007 in India, 2005-2007 in South Korea, 2005-2007 in Taiwan, and 2001-2003 in Thailand; the numbers of rotavirus-related deaths in each country; and published rotavirus serotype-specific efficacy of RV5. The model projected an overall effectiveness in the region of 82% to 89% against RGE-related hospitalizations and a substantial reduction in RGE-related deaths, suggesting that RV5 could substantially reduce the burden of rotavirus disease in Asia.


Assuntos
Hospitalização/estatística & dados numéricos , Infecções por Rotavirus/epidemiologia , Infecções por Rotavirus/prevenção & controle , Vacinas contra Rotavirus/administração & dosagem , Vacinas contra Rotavirus/imunologia , Análise de Sobrevida , Ásia/epidemiologia , Pré-Escolar , Gastroenterite/epidemiologia , Gastroenterite/mortalidade , Gastroenterite/prevenção & controle , Humanos , Lactente , Recém-Nascido , Modelos Estatísticos , Infecções por Rotavirus/mortalidade , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia
14.
Hum Vaccin ; 7(5): 563-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21441783

RESUMO

Post-hoc analyses of the Rotavirus Efficacy and Safety Trial (REST) were conducted to determine whether the pentavalent rotavirus vaccine (RV5) confers early protection against rotavirus gastroenteritis (RVGE) before completion of the 3-dose regimen. To evaluate the efficacy of RV5 between doses in reducing the rates of RVGE-related hospitalizations and emergency department (ED) visits in infants who ultimately received all 3 doses of RV5/placebo, events occurring from 2 weeks after the first and second doses to receipt of the subsequent dose (Analysis A) and events occurring from 2 weeks after the first and second doses to 2 weeks after the subsequent dose (Analysis B) were analyzed. In Analysis A, RV5 reduced the rates of combined hospitalizations and ED visits for G1-G4 RVGE or RVGE regardless of serotype between doses 1 and 2 by 100% (95% confidence interval [CI]: 72-100%) or 82% (95% CI: 39-97%), respectively, and between doses 2 and 3, RV5 reduced the rates of combined hospitalizations and ED visits for G1-G4 RVGE or RVGE regardless of serotype by 91% (95% CI: 63-99%) or 84% (95% CI: 54-96%), respectively. Similar rate reductions were observed in Analysis B. These data suggest that RV5 provides a high level of protection between doses against hospitalizations and ED visits for RVGE starting as early as 14 days after the first dose.


Assuntos
Gastroenterite/prevenção & controle , Imunização Secundária/métodos , Infecções por Rotavirus/prevenção & controle , Vacinas contra Rotavirus/administração & dosagem , Vacinas contra Rotavirus/imunologia , Vacinação/métodos , Gastroenterite/epidemiologia , Gastroenterite/imunologia , Hospitalização/estatística & dados numéricos , Humanos , Imunização Secundária/efeitos adversos , Lactente , Placebos/administração & dosagem , Infecções por Rotavirus/epidemiologia , Infecções por Rotavirus/imunologia , Vacinas contra Rotavirus/efeitos adversos , Vacinação/efeitos adversos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/imunologia
15.
Mem Inst Oswaldo Cruz ; 106(5): 541-5, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21894373

RESUMO

RotaTeq® (Merck & Company, Inc, Whitehouse Station, NJ, USA) is an oral pentavalent rotavirus vaccine (RV5) that has shown high and consistent efficacy in preventing rotavirus gastroenteritis (RGE) in randomised clinical trials previously conducted in industrialised countries with high medical care resources. To date, the efficacy and effectiveness data for RV5 are available in some Latin American countries, but not Brazil. In this analysis, we projected the effectiveness of RV5 in terms of the percentage reduction in RGE-related hospitalisations among children less than five years of age in four regions of Brazil, using a previously validated mathematical model. The model inputs included hospital-based rotavirus surveillance data from Goiânia, Porto Alegre, Salvador and São Paulo from 2005-2006, which provided the proportions of rotavirus attributable to serotypes G1, G2, G3, G4 and G9, and published rotavirus serotype-specific efficacy from the Rotavirus Efficacy and Safety Trial. The model projected an overall percentage reduction of 93% in RGE-related hospitalisations, with an estimated annual reduction in RGE-related hospitalisations between 42,991-77,383 in the four combined regions of Brazil. These results suggest that RV5 could substantially prevent RGE-related hospitalisations in Brazil.


Assuntos
Gastroenterite/prevenção & controle , Infecções por Rotavirus/prevenção & controle , Vacinas contra Rotavirus/uso terapêutico , Adolescente , Brasil , Criança , Pré-Escolar , Feminino , Gastroenterite/virologia , Hospitalização , Humanos , Lactente , Masculino , Modelos Estatísticos , Avaliação de Programas e Projetos de Saúde , Vacinas Atenuadas/uso terapêutico
16.
J Infect Dis ; 202 Suppl: S87-92, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20684723

RESUMO

Phase III studies of an oral, live, pentavalent, human-bovine reassortant rotavirus vaccine (RotaTeq; Merck) in developed countries have demonstrated that it is well tolerated with regard to intussusception and other adverse events and is efficacious in preventing rotavirus gastroenteritis and associated healthcare encounters. However, it cannot be assumed that rotavirus vaccines will be equally efficacious in infants and young children in the developing world. Differences in host populations, associated health conditions, and the epidemiology of rotavirus disease could affect vaccine performance. Concern about the potential for differences in efficacy stems from studies of previous candidate rotavirus vaccines, including bovine and rhesus rotaviruses, which showed no or variable efficacy in developing regions. Given this history, the World Health Organization (WHO) recommended that the efficacy of "new" rotavirus vaccines should be demonstrated in diverse geographic areas, including developing countries, before widespread implementation. Successful implementation of any rotavirus vaccine in the developing world requires additional clinical research and sharing of early introduction experiences. We discuss efforts to bring RotaTeq vaccine to the developing world. Critical steps to achieve this goal include the clinical evaluation of vaccine safety and efficacy in a multisite trial in Asia and Africa, evaluation of concomitant use with other pediatric vaccines routinely used, and vaccine assessment in special populations (premature, human immunodeficiency virus-infected, and malnourished infants). Completion of WHO prequalification of RotaTeq and affordability are also key requirements to routine vaccine introduction. The RotaTeq Partnership with the Nicaraguan Ministry of Health provides an example of the successful introduction of this vaccine into a developing world country.


Assuntos
Países em Desenvolvimento , Infecções por Rotavirus/prevenção & controle , Vacinas contra Rotavirus , Ensaios Clínicos como Assunto , Países Desenvolvidos , Surtos de Doenças , Infecções por HIV/complicações , Humanos , Programas de Imunização , Esquemas de Imunização , Lactente , Transtornos da Nutrição do Lactente/complicações , Recém-Nascido , Recém-Nascido Prematuro , Nicarágua/epidemiologia , Infecções por Rotavirus/epidemiologia , Vacinas contra Rotavirus/administração & dosagem , Vacinas contra Rotavirus/efeitos adversos , Vacinas contra Rotavirus/normas
17.
Emerg Infect Dis ; 16(4): 625-30, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20350376

RESUMO

G11 rotaviruses are believed to be of porcine origin. However, a limited number of G11 rotaviruses have been recently isolated from humans in combination with P[25], P[8], P[6], and P[4]. To investigate the evolutionary relationships of these strains, we analyzed the complete genomes of 2 human G11P[25] strains, 2 human G11P[8] strains, and 3 porcine reference strains. Most of the 11 gene segments of these 7 strains belonged to genotype 1 (Wa-like). However, phylogenetic clustering patterns suggested that an unknown G11P[25] strain with a new I12 VP6 genotype was transmitted to the human population, in which it acquired human genotype 1 gene segments through reassortment, resulting in a human G11P[8] rotavirus strain with an entire human Wa-genogroup backbone. This Wa-like backbone is believed to have caused the worldwide spread of human G9 and G12 rotaviruses. G11 human rotavirus strains should be monitored because they may also become major human pathogens.


Assuntos
Genes Virais/genética , Vírus Reordenados/genética , Infecções por Rotavirus/virologia , Rotavirus/genética , Animais , Genótipo , Humanos , Filogenia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Suínos/virologia , Doenças dos Suínos/virologia
18.
J Virol ; 83(7): 2917-29, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19153225

RESUMO

A limited number of human G6P[14] rotavirus strains that cause gastroenteritis in humans have been isolated in Europe and Australia. The complete genome sequences were determined for five of these human strains--B10925-97 (isolated in Belgium in 1997), 111/05-27 (Italy, 2005), PA169 (Italy, 1987), MG6 (Australia, 1993), and Hun5 (Hungary, 1997)--and their genetic relatedness to animal rotavirus strains was evaluated by sequencing the complete genome of the sheep rotavirus OVR762 (G8P[14]; Spain, 2002), the guanaco (Lama guanicoe) rotavirus strains Arg/Chubut/99 and Arg/Río Negro/98 (G8P[14] and G8P[1], respectively; Argentina, 1999 and 1998), the sable antelope strain RC-18/08 (G6P[14]; South Africa, 2008), and the bovine rotavirus strain Arg/B383/98 (G15P[11]; Argentina, 1998). These analyses revealed an overall consensus genomic constellation (G6/G8)-P[14]-I2-(R2/R5)-C2-M2-(A3/A11)-N2-T6-(E2/E12)-H3, together with a few gene reassortments, and the phylogenetic analyses confirmed that the P[14] human strains evaluated in this study were closely related to rotavirus strains isolated from sheep, cattle, goats, guanacos, and antelopes and to rabbits (albeit to a lesser extent), suggesting that one (or more) of these animal species might be the source of the human G6P[14] strains. The main feature of the genotype and phylogenetic analyses was the close overall genomic relatedness between the five human G6P[14] rotavirus strains and the ovine and antelope rotavirus strains. Taken together, these data strongly suggest a common origin for the human P[14] strains and those of the even-toed ungulates belonging to the mammalian order Artiodactyla, with sheep probably playing a key role in the interspecies transmission responsible for the introduction of P[14] rotavirus strains into the human population.


Assuntos
Gastroenterite/virologia , RNA Viral/genética , Infecções por Rotavirus/virologia , Rotavirus/genética , Rotavirus/isolamento & purificação , Austrália , Pré-Escolar , Europa (Continente) , Evolução Molecular , Humanos , Lactente , Dados de Sequência Molecular , Filogenia , Análise de Sequência de DNA , Homologia de Sequência
19.
Eur J Pediatr ; 169(11): 1379-86, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20559656

RESUMO

Rotavirus Efficacy and Safety Trial (REST) enrolled nearly 70,000 infants, of whom more than 23,000 were from Finland. REST determined the efficacy of the pentavalent rotavirus vaccine (RV5) against rotavirus-related hospitalisations and emergency department (ED) visits in the first year after vaccination. Finnish infants initially in REST transitioned into the Finnish Extension Study (FES), where they were followed for rotavirus-related hospitalisations and ED visits through their second year of life and beyond. FES identified 150 (31%) additional rotavirus gastroenteritis (RVGE) cases beyond those identified in REST in the Finnish participants. Overall, RV5 reduced RVGE hospitalisations and ED visits, regardless of the rotavirus serotype, by 93.8% (95% confidence interval [CI]: 90.8-95.9%) for up to 3.1 years following the last vaccine dose. Vaccine efficacy against combined hospitalisations and ED visits between ages 4 months to 11 months, 12 months to 23 months, and 24 months to 35 months was 93.9% (95% CI: 89.1-96.9%), 94.4% (95% CI: 90.2-97.0%), and 85.9% (95% CI: 51.6-97.2%), respectively. The reduction of hospitalisations and ED visits due to any acute gastroenteritis, rotavirus or not, was 62.4% (95% CI: 57.6-66.6%) over the entire follow-up period. The results from FES confirm that RV5 induces high and sustained protection against rotavirus-related hospitalisations and ED visits, and has a very substantial impact on all gastroenteritis-related hospitalisations and ED visits into the third year of life in Finnish children.


Assuntos
Gastroenterite/prevenção & controle , Hospitalização/tendências , Infecções por Rotavirus/prevenção & controle , Vacinas contra Rotavirus/administração & dosagem , Doença Aguda , Anticorpos Antivirais/imunologia , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Finlândia/epidemiologia , Seguimentos , Gastroenterite/epidemiologia , Gastroenterite/virologia , Humanos , Incidência , Lactente , Masculino , Estudos Retrospectivos , Rotavirus/imunologia , Infecções por Rotavirus/epidemiologia , Infecções por Rotavirus/virologia , Resultado do Tratamento , Vacinas Atenuadas/administração & dosagem
20.
Vaccine ; 38(3): 578-587, 2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31679865

RESUMO

OBJECTIVE: To assess the safety and immunogenicity of the MF59®-adjuvanted trivalent influenza vaccine (aTIV; Fluad®) compared with modified aTIV formulations. METHODS: A total of 196 subjects ≥ 65 years were randomized to receive7different formulations of vaccine containing a range of adjuvant and antigen dosesby single injection, or divided into two injections at a single time point. The primary study objective was to compare the serologic response of different formulations of aTIV containing increased amounts of adjuvant and antigen21 days after vaccination. Subjects were followed for immunogenicity and safety for one year. RESULTS: The highest immune response, as measured by hemagglutination inhibition (HI) assay, 3 weeks after vaccination was observed in subjects in Group 6 with GMT 382.2 (95% confidence interval [CI] 237.5 to 615.0), 552.3 (364.8 to 836.1), and 54.1 (36.9 to 79.4) against A/H1N1, A/H3N2, and B respectively. Rates of seroconversion were also generally highest in this treatment group: 75% (95% CI 55.1 to 89.3), 75% (55.1 to 89.3), and 42.9% (24.5 to 62.8), respectively, against A/H1N1, A/H3N2, and B strains. The highest incidence of solicited adverse events (AEs) was reported by subjects who received both the highest dosage of antigen in combination with the highest dosage of adjuvant at the same site: 67.9% and 57.1% in Groups 4 and 6, respectively. The majority of solicited AEs were mild to moderate in severity. The number of unsolicited AEs was similar across the different dosages. CONCLUSION: In this phase I trial of adults ≥ 65 years of age who received increased adjuvant and antigen dosages relative to the licensed aTIV, increased dosage of MF59 resulted in increased immunogenicity against all 3 components of seasonal influenza vaccine. The increase in immunogenicity was accompanied by an increase in the incidence of local reactogenicity.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos Virais/administração & dosagem , Imunogenicidade da Vacina , Vacinas contra Influenza/administração & dosagem , Influenza Humana/epidemiologia , Polissorbatos/administração & dosagem , Esqualeno/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos , Idoso , Idoso de 80 Anos ou mais , Antígenos Virais/efeitos adversos , Antígenos Virais/imunologia , Formas de Dosagem , Relação Dose-Resposta Imunológica , Feminino , Alemanha/epidemiologia , Humanos , Imunogenicidade da Vacina/imunologia , Vacinas contra Influenza/efeitos adversos , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Masculino , Polissorbatos/efeitos adversos , Método Simples-Cego , Esqualeno/efeitos adversos , Esqualeno/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA