Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
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
2.
JBI Evid Synth ; 19(6): 1263-1291, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33741840

RESUMO

OBJECTIVE: The objective of this review was to determine the diagnostic accuracy of [-2]proPSA (p2PSA) and the Prostate Health Index compared to the Gleason score in determining the aggressiveness of prostate cancer. INTRODUCTION: Prostate cancer is the most commonly diagnosed cancer in men. However, the utility of currently available biomarkers for determining the aggressive form of the disease remains unknown. This review sought to determine the diagnostic accuracy of two new biomarkers in determining the aggressive form of prostate cancer. INCLUSION CRITERIA: Diagnostic accuracy studies that enrolled men of any age and any prostate specific antigen (PSA) level with histologically confirmed prostate cancer in which Prostate Health Index and p2PSA were assessed in comparison to Gleason score for the determination of aggressive prostate cancer were considered for inclusion. There was no time limitation on study inclusion. METHODS: A three-step search strategy was utilized to identify both published and unpublished studies in the English language in the following sources: PubMed, Cochrane Central Register of Controlled Trials, CINAHL, Web of Science, Google Scholar, MedNar, and SIGLE. Databases were searched from inception to January 2019. Study selection, critical appraisal, data extraction, and data synthesis were done according to the approach recommended by JBI. RESULTS: A total of 12 studies (n = 8462) that recruited men with aggressive prostate cancer were considered in this review. The majority of included subjects had a total PSA level of 2 to 10ng/mL. The sensitivity of the Prostate Health Index ranged from 67% to 97% while specificity ranged from 6% to 64%. At a Prostate Health Index threshold of 25 and below (three studies, n = 3222), pooled sensitivity was 97% (95% confidence interval [CI], 95% to 98%) and specificity was 10% (95% CI, 6% to 16%). At a Prostate Health Index threshold of between 26 and 35 (six studies, n = 6030), pooled sensitivity was 87% (95% CI, 8% to 91%) and specificity was 45% (95% CI, 39% to 50%). At a Prostate Health Index threshold of 36 and above (five studies, n = 1476), pooled sensitivity was 72% (95% CI, 64% to 79%) and specificity was 74% (95% CI, 68% to 80%). Only one study assessed p2PSA. Sensitivity ranged from 80% to 95%, and specificity ranged from 9.9% to 27.9% with increasing threshold values from 7.9 to 10.9ng/mL. CONCLUSIONS: Overall, both Prostate Health Index and p2PSA have acceptable accuracy for the determination of the likelihood of aggressive prostate cancer. However, the inverse relationship between sensitivity and specificity makes it difficult to determine an optimum cut-off value for positivity. Further research is warranted to determine their utility in the management of prostate cancer.


Assuntos
Antígeno Prostático Específico , Neoplasias da Próstata , Humanos , Masculino , Gradação de Tumores , Neoplasias da Próstata/diagnóstico , Sensibilidade e Especificidade
4.
Vaccine ; 30 Suppl 1: A52-60, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22520137

RESUMO

BACKGROUND: Rotavirus gastroenteritis (RVGE) is a leading cause of death in African children. The efficacy of pentavalent rotavirus vaccine (PRV) against severe RVGE evaluated in Ghana, Kenya, and Mali in a randomized, double-blind, placebo-controlled trial, showed a combined regional efficacy of 39.3% (95% confidence interval [CI]: 19.1,54.7) in nearly 2 years of follow-up. This report concentrates on the Kenya findings. METHODS: Infants received 3 doses of PRV/placebo at approximately 6-, 10-, and 14-weeks of age. HIV testing was offered to all participants. Data on illness symptoms and signs were collected upon presentation to healthcare facilities, where stools were collected, and analyzed by rotavirus-specific enzyme-linked immunosorbent assay. The primary endpoint was severe RVGE (Vesikari score ≥ 11), occurring ≥ 14 days following the third dose. At monthly home visits, symptoms of illnesses during the past 2 weeks were solicited and limited physical exams were performed; dehydration was defined by WHO's Integrated Management of Childhood Illness. FINDINGS: Vaccine efficacy (VE) against severe RVGE through nearly 2 years of follow-up among 1308 Kenyan children was 63.9% (95% CI: -5.9,89.8). Through the first year of life, VE against severe RVGE was 83.4% (95% CI: 25.5,98.2). From home visits, VE against all-cause gastroenteritis with severe dehydration was 34.4% (95% CI: 5.3,54.6) through the first year and 29.7% (95% CI: 2.5,49.3) through the entire follow-up period. The reduction in incidence of gastroenteritis with severe dehydration in the community during the first year of life (19.0 cases/100 person-years) was almost six times greater than the reduction in severe RVGE presenting to the clinic (3.3/100 person-years). Oral rehydration solution use was lower among PRV recipients (VE 23.1%, 95% CI: 8.8,35.1). An estimated 41% of gastroenteritis with severe dehydration in the first year reported at home was rotavirus-related. CONCLUSIONS: PRV significantly reduced severe RVGE in Kenya. The impact of PRV might be greatest in rural Africa in protecting the many children who develop severe gastroenteritis and cannot access health facilities.


Assuntos
Gastroenterite/prevenção & controle , Infecções por Rotavirus/prevenção & controle , Vacinas contra Rotavirus/administração & dosagem , Vacinas contra Rotavirus/imunologia , Vacinação/métodos , Administração Oral , Método Duplo-Cego , Fezes/virologia , Feminino , Gastroenterite/epidemiologia , Gastroenterite/patologia , Genótipo , Infecções por HIV/complicações , Infecções por HIV/imunologia , Humanos , Incidência , Lactente , Quênia/epidemiologia , Masculino , Placebos/administração & dosagem , Rotavirus/classificação , Rotavirus/genética , Rotavirus/isolamento & purificação , Infecções por Rotavirus/epidemiologia , Infecções por Rotavirus/patologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia
5.
Vaccine ; 30 Suppl 1: A61-70, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22520138

RESUMO

Two multicenter Phase III trials were conducted in five countries from March 2007 to March 2009 to evaluate the safety and efficacy of the pentavalent rotavirus vaccine (PRV), RotaTeq(®), in Africa and Asia. In this report, we evaluate the safety of this vaccine, including among HIV-infected and HIV-exposed infants, in Kenya. 1308 Infants were randomized 1:1 to receive 3 doses of PRV/placebo at approximately 6, 10, and 14 weeks of age. HIV counseling and testing were offered to all participants. A positive PCR result indicated HIV infection; the presence of HIV antibody in PCR-negative children indicated HIV exposure without HIV infection. All serious adverse events (SAE) within 14 days of any dose, and vaccine-related SAEs, intussusception, and deaths occurring at any time during the study, were reported ("SAE surveillance"). In addition, 297 participants were followed for 42 days after any dose for any adverse event (AE), regardless of severity ("intensive safety surveillance"). The safety evaluation was stratified by HIV status. SAEs were reported in 20/649 vaccine recipients (3.1%) and 21/643 placebo recipients (3.3%) within 14 days following vaccination (p = 0.9). The most common SAE in the vaccinated group was pneumonia (1.7%). No individual SAE was significantly more common among vaccine vs. placebo recipients. Seventy-two deaths were reported, 38 (5.9%) and 34 (5.3%) among vaccine and placebo recipients, respectively (p = 0.66). No cases of intussusception were reported. During intensive safety surveillance, 137/147 (93.2%) vaccine recipients and 147/150 (98.0%) placebo recipients experienced one or more AEs (risk ratio = 0.95; 95% CI: 0.91-1.0; p = 0.05). 88.5% of the infants were tested for HIV infection; 21/581 (3.6%) children in the vaccine group and 17/577 (2.9%) in the placebo group were HIV-infected. Among the 37 HIV-infected infants with full safety follow-up, 5/21 (23.8%) vaccine recipients and 2/16 (12.5%) placebo recipients reported an SAE (p = 0.67). In total, 12 deaths occurred among identified HIV-infected infants: 8 (38%) receiving vaccine vs. 4 (23.5%) receiving placebo (RR = 1.6, 95% CI: 0.59-4.5). Among the 21 HIV-infected infants in the vaccine group, 2 of 8 deaths were gastroenteritis-related; among the 17 HIV-infected infants in the placebo group, 3 of 4 deaths were gastroenteritis-related. There were no significant differences in serious or non-serious AEs, including vaccine-related SAEs, between the 88 HIV-exposed vaccine recipients vs. the 89 HIV-exposed placebo recipients. PRV appears to be a safe intervention against rotavirus gastroenteritis among infants in Kenya. AEs, including serious AEs, were not associated with receipt of vaccine. Further, SAEs were not significantly more common among HIV-infected or HIV-exposed participants; however, the low number of HIV-infected infants did not provide sufficient power to fully assess safety in HIV-infected vaccine recipients.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Gastroenterite/prevenção & controle , Infecções por Rotavirus/prevenção & controle , Vacinas contra Rotavirus/administração & dosagem , Vacinas contra Rotavirus/efeitos adversos , Vacinação/efeitos adversos , Vacinação/métodos , Administração Oral , Método Duplo-Cego , Feminino , Gastroenterite/epidemiologia , HIV/isolamento & purificação , Anticorpos Anti-HIV/sangue , Infecções por HIV/complicações , Infecções por HIV/imunologia , Humanos , Lactente , Recém-Nascido , Intussuscepção/induzido quimicamente , Intussuscepção/epidemiologia , Quênia/epidemiologia , Masculino , Placebos/administração & dosagem , Infecções por Rotavirus/epidemiologia , Vacinas contra Rotavirus/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/imunologia
6.
Vaccine ; 30 Suppl 1: A86-93, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22520142

RESUMO

We recently completed a double-blind, placebo-controlled, multicenter Phase III clinical trial of the pentavalent rotavirus vaccine (PRV) in three African countries, Ghana, Kenya, and Mali, from April 2007 to March 2009. The immunogenicity of PRV in African infants is described. In total, 5468 infants were randomized 1:1 to receive 3 doses of PRV or placebo at approximately 6, 10, and 14 weeks of age. Breastfeeding and concomitant administration of EPI vaccines, including OPV, were allowed, and HIV-infected infants were not excluded. Immunogenicity of PRV was assessed by measuring serum anti-rotavirus IgA responses, as well as serum neutralization antibody (SNA) to the human rotavirus serotypes G1, G2, G3, G4 and P1A[8] in approximately 150 infants per country. Sera were collected pre-dose 1 (pD1) and approximately 14 days post-dose 3 (PD3) for immunological analysis. For the sero-response rates (≥ 3-fold rise from pD1 to PD3), the number of subjects evaluable included those with both pD1 and PD3 data available. PRV was immunogenic in African children and significantly reduced severe RVGE in African children through the first two years of life. The pooled anti-rotavirus IgA sero-response rate was 78.3%, with consistent rates in each of the African sites: 73.8% (Kenya), 78.9% (Ghana), and 82.5% (Mali); but generally lower than that reported in Europe and USA. PD3 GMTs (28.2 dilution-units) were 5-10 times lower than those assessed in subjects in clinical trials in developed countries. SNA responses to human rotavirus serotypes G1-G4 and P1A[8] ranged from 6.3% (G3) to 26.5% (G4). PD3 SNA GMTs to G1 and P1A[8] were 4-fold and 3-fold lower respectively, when compared to the corresponding GMTs in subjects who received PRV in similar studies conducted in developed countries. PRV was immunogenic in African infants, and the anti-rotavirus IgA sero-response rates were similar across all three African sites although lower than those observed in Europe and USA. While immune correlates of protection have not been established for rotavirus, the findings are consistent with lower efficacy rates demonstrated during this trial. Further investigation is needed to understand the reason for the lower immunogenicity observed.


Assuntos
Gastroenterite/prevenção & controle , Infecções por Rotavirus/prevenção & controle , Vacinas contra Rotavirus/administração & dosagem , Vacinas contra Rotavirus/imunologia , Vacinação/métodos , Administração Oral , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Método Duplo-Cego , Feminino , Gastroenterite/epidemiologia , Gana/epidemiologia , Humanos , Imunoglobulina A/sangue , Lactente , Quênia/epidemiologia , Masculino , Mali/epidemiologia , Placebos/administração & dosagem , Infecções por Rotavirus/epidemiologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA