Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Trop Med Hyg ; 101(1): 164-179, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31115304

RESUMEN

The CYD-TDV vaccine is licensed in multiple endemic countries based on vaccine efficacy (VE) against symptomatic, virologically confirmed dengue demonstrated in two phase 3 trials (CYD14, 2- to 14-year-olds, Asia; CYD15, 9- to 16-year-olds, Latin America). 50% plaque reduction neutralization test (PRNT50) titers at baseline and month 13 (post-vaccination) were associated with VE and may enable bridging VE to adults. Two phase 2 trials of CYD-TDV measured baseline and month 13 PRNT50 titers: CYD22 (9- to 45-year-olds, Vietnam) and CYD47 (18- to 45-year-olds, India). 50% plaque reduction neutralization test distributions were compared between age cohorts, and four versions of an epidemiological bridging method were used to estimate VE against any serotype (dengue virus [DENV]-Any) and against each serotype over 25 months post first vaccination in a hypothetical CYD14 + CYD15 18- to 45-year-old cohort (bridging population 1) and in the actual CYD47 18- to 45-year-old cohort (bridging population 2). Baseline and month 13 geometric mean PRNT50 titers to each serotype were significantly greater in 18- to 45-year-olds than 9- to 16-year-olds for all comparisons. The four methods estimated VE against DENV-Any at 75.3-86.0% (95% CIs spanning 52.5-100%) for bridging population 1 and 68.4-77.5% (95% CIs spanning 42.3-88.5%) for bridging population 2. The vaccine efficacy against serotype 1, 2, 3, and 4 was estimated at 56.9-76.9%, 68.3-85.8%, 91.4-95.0%, and 93.2-100% (bridging population 1) and 44.5-66.9%, 53.2-69.2%, 79.8-92.0%, and 90.6-95.0% (bridging population 2), respectively; thus, CYD-TDV would likely confer improved efficacy in adults than 9- to 16-year-olds. Using the same methods, we predicted VE against hospitalized DENV-Any over 72 months of follow-up, with estimates 59.1-73.5% (95% CIs spanning 40.9-92.2%) for bridging population 1 and 50.9-65.9% (95% CIs spanning 38.1-82.1%) for bridging population 2.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Vacunas contra el Dengue/normas , Virus del Dengue/inmunología , Dengue/prevención & control , Enfermedades Endémicas/prevención & control , Adolescente , Adulto , Niño , Vacunas contra el Dengue/inmunología , Virus del Dengue/clasificación , Humanos , Persona de Mediana Edad , Serogrupo , Ensayo de Placa Viral , Adulto Joven
4.
Biometrics ; 74(4): 1271-1281, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29701875

RESUMEN

A common scientific problem is to determine a surrogate outcome for a long-term outcome so that future randomized studies can restrict themselves to only collecting the surrogate outcome. We consider the setting that we observe n independent and identically distributed observations of a random variable consisting of baseline covariates, a treatment, a vector of candidate surrogate outcomes at an intermediate time point, and the final outcome of interest at a final time point. We assume the treatment is randomized, conditional on the baseline covariates. The goal is to use these data to learn a most-promising surrogate for use in future trials for inference about a mean contrast treatment effect on the final outcome. We define an optimal surrogate for the current study as the function of the data generating distribution collected by the intermediate time point that satisfies the Prentice definition of a valid surrogate endpoint and that optimally predicts the final outcome: this optimal surrogate is an unknown parameter. We show that this optimal surrogate is a conditional mean and present super-learner and targeted super-learner based estimators, whose predicted outcomes are used as the surrogate in applications. We demonstrate a number of desirable properties of this optimal surrogate and its estimators, and study the methodology in simulations and an application to dengue vaccine efficacy trials.


Asunto(s)
Biomarcadores , Biometría/métodos , Simulación por Computador/estadística & datos numéricos , Ensayos Clínicos Controlados Aleatorios como Asunto/estadística & datos numéricos , Vacunas contra el Dengue/normas , Humanos , Funciones de Verosimilitud , Evaluación de Resultado en la Atención de Salud
5.
Math Biosci ; 294: 15-32, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28935561

RESUMEN

In this paper we study a mathematical model to analyse the optimal vaccination age against Dengue in Brazil. Data from Brazil are used to estimate the basic reproduction numbers for each of the four Dengue serotypes and then the optimal vaccination age is calculated using a method due to Hethcote [1]. The vaccine has different efficacies against each serotype. Vaccination that is too early is ineffective as individuals are protected by maternal antibodies but leaving vaccination until later may allow the disease to spread. First of all the optimal vaccination ages are calculated where there is just one serotype in circulation and then when there are multiple serotypes. The calculations are done using data both assuming constant vaccine efficacy and age-dependent vaccine efficacy against a given serotype. The multiple serotype calculations are repeated assuming that the first infection is a risky infection and that it is not (to model Dengue Antibody Enhancement). The calculations are then repeated when any third or fourth Dengue infections are asymptomatic, so that two Dengue infections with different serotypes provide effective permanent immunity. The calculations are also repeated when the age-dependent risk function (fitted to Brazilian data) is hospitalisation from Dengue and when it is mortality due to Dengue. We find a wide variety of optimal vaccination ages depending on both the serotypes in circulation and the assumptions of the model.


Asunto(s)
Vacunas contra el Dengue/normas , Dengue/prevención & control , Modelos Teóricos , Vacunación/normas , Factores de Edad , Brasil , Humanos
7.
PLoS Negl Trop Dis ; 10(12): e0005179, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-28002420

RESUMEN

BACKGROUND: With approximately 3 billion people at risk of acquiring the infection, dengue fever is now considered the most important mosquito-borne viral disease in the world, with 390 million dengue infections occurring every year, of which 96 million manifest symptoms with any level of disease severity. Treatment of uncomplicated dengue cases is only supportive and severe dengue cases require hospital intensive care. A vaccine now licensed in several countries and developed by Sanofi Pasteur (CYD-TDV, named Dengvaxia), was able to protect, in the first 25 months of the two Phase III, 66% of a subset of 9-16 year old participants. However, a significantly lower efficacy (including negative vaccine efficacy) was noted for children younger than 9 years of age. METHODOLOGY/PRINCIPAL FINDINGS: Analysis of year 3 results of phase III trials of Dengvaxia suggest high rates of protection of vaccinated partial dengue immunes but high rates of hospitalizations during breakthrough dengue infections of persons who were vaccinated when seronegative, with vaccine appearing to induce enhancing antibodies (ADE). An age structured model was developed based on Sanofi's recommendation to vaccinate persons age 945 years in dengue endemic countries. The model was used to explore the clinical burden of two vaccination strategies: 1) Vaccinate 4 or 20% of individuals, ages 9-45 years, seropositives and seronegatives, and 2) vaccinate 4 or 20% of individuals, ages 9-45 years, who are dengue immune only. CONCLUSIONS/SIGNIFICANCE: Our results show that vaccinating dengue monotypic immune individuals prevents dengue hospitalizations, but at the same time dengue infections of vaccine-sensitized persons increases hospitalizations. When the vaccine is given only to partial immune individuals, after immunological screening of the population, disease burden decreases considerably.


Asunto(s)
Vacunas contra el Dengue/efectos adversos , Vacunas contra el Dengue/inmunología , Dengue/inmunología , Dengue/prevención & control , Hospitalización/estadística & datos numéricos , Vacunación/estadística & datos numéricos , Adolescente , Adulto , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Niño , Ensayos Clínicos Fase III como Asunto , Dengue/epidemiología , Dengue/virología , Vacunas contra el Dengue/administración & dosificación , Vacunas contra el Dengue/normas , Virus del Dengue/inmunología , Enfermedades Endémicas/prevención & control , Enfermedades Endémicas/estadística & datos numéricos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Dengue Grave/inmunología , Dengue Grave/prevención & control , Dengue Grave/virología , Vacunación/legislación & jurisprudencia , Vacunación/normas , Vacunas Atenuadas/inmunología , Adulto Joven
8.
PLoS Med ; 13(11): e1002181, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27898668

RESUMEN

BACKGROUND: Large Phase III trials across Asia and Latin America have recently demonstrated the efficacy of a recombinant, live-attenuated dengue vaccine (Dengvaxia) over the first 25 mo following vaccination. Subsequent data collected in the longer-term follow-up phase, however, have raised concerns about a potential increase in hospitalization risk of subsequent dengue infections, in particular among young, dengue-naïve vaccinees. We here report predictions from eight independent modelling groups on the long-term safety, public health impact, and cost-effectiveness of routine vaccination with Dengvaxia in a range of transmission settings, as characterised by seroprevalence levels among 9-y-olds (SP9). These predictions were conducted for the World Health Organization to inform their recommendations on optimal use of this vaccine. METHODS AND FINDINGS: The models adopted, with small variations, a parsimonious vaccine mode of action that was able to reproduce quantitative features of the observed trial data. The adopted mode of action assumed that vaccination, similarly to natural infection, induces transient, heterologous protection and, further, establishes a long-lasting immunogenic memory, which determines disease severity of subsequent infections. The default vaccination policy considered was routine vaccination of 9-y-old children in a three-dose schedule at 80% coverage. The outcomes examined were the impact of vaccination on infections, symptomatic dengue, hospitalised dengue, deaths, and cost-effectiveness over a 30-y postvaccination period. Case definitions were chosen in accordance with the Phase III trials. All models predicted that in settings with moderate to high dengue endemicity (SP9 ≥ 50%), the default vaccination policy would reduce the burden of dengue disease for the population by 6%-25% (all simulations: -3%-34%) and in high-transmission settings (SP9 ≥ 70%) by 13%-25% (all simulations: 10%- 34%). These endemicity levels are representative of the participating sites in both Phase III trials. In contrast, in settings with low transmission intensity (SP9 ≤ 30%), the models predicted that vaccination could lead to a substantial increase in hospitalisation because of dengue. Modelling reduced vaccine coverage or the addition of catch-up campaigns showed that the impact of vaccination scaled approximately linearly with the number of people vaccinated. In assessing the optimal age of vaccination, we found that targeting older children could increase the net benefit of vaccination in settings with moderate transmission intensity (SP9 = 50%). Overall, vaccination was predicted to be potentially cost-effective in most endemic settings if priced competitively. The results are based on the assumption that the vaccine acts similarly to natural infection. This assumption is consistent with the available trial results but cannot be directly validated in the absence of additional data. Furthermore, uncertainties remain regarding the level of protection provided against disease versus infection and the rate at which vaccine-induced protection declines. CONCLUSIONS: Dengvaxia has the potential to reduce the burden of dengue disease in areas of moderate to high dengue endemicity. However, the potential risks of vaccination in areas with limited exposure to dengue as well as the local costs and benefits of routine vaccination are important considerations for the inclusion of Dengvaxia into existing immunisation programmes. These results were important inputs into WHO global policy for use of this licensed dengue vaccine.


Asunto(s)
Vacunas contra el Dengue/economía , Vacunas contra el Dengue/normas , Modelos Teóricos , Salud Pública , Seguridad , Vacunación/métodos , Niño , Análisis Costo-Beneficio , Vacunas contra el Dengue/efectos adversos , Humanos , Estudios Seroepidemiológicos , Vacunación/efectos adversos , Vacunación/economía , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/economía , Vacunas Atenuadas/normas , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/economía , Vacunas Sintéticas/normas
9.
PLoS Med ; 13(11): e1002182, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27898675

RESUMEN

In a Perspective, Jacqueline Deeen discusses challenges in balancing the individual and population risks and benefits for CYD-TDV (Dengvaxia), the first available dengue vaccine.


Asunto(s)
Vacunas contra el Dengue/normas , Dengue/prevención & control , Vacunación , Vacunas contra el Dengue/efectos adversos , Humanos , Medición de Riesgo , Vacunación/normas , Vacunación/estadística & datos numéricos
10.
Am J Trop Med Hyg ; 94(6): 1348-1358, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27022153

RESUMEN

We evaluated the safety and immunogenicity of two doses of a live-attenuated, tetravalent dengue virus vaccine (F17/Pre formulation) and a booster dose in a dengue endemic setting in two studies. Seven children (7- to 8-year-olds) were followed for 1 year after dose 2 and then given a booster dose (F17/Pre formulation), and followed for four more years (Child study). In the Infant study, 49 2-year-olds, vaccinated as infants, were followed for approximately 3.5 years after dose 2 and then given a booster dose (F17) and followed for one additional year. Two clinically notable events were observed, both in dengue vaccine recipients in the Infant study: 1 case of dengue approximately 2.7 years after dose 2 and 1 case of suspected dengue after booster vaccinations. The booster vaccinations had a favorable safety profile in terms of reactogenicity and adverse events reported during the 1-month follow-up periods. No vaccine-related serious adverse events were reported during the studies. Neutralizing antibodies against dengue viruses 1-4 waned during the 1-3 years before boosting, which elicited a short-lived booster response but did not provide a long-lived, multivalent antibody response in most subjects. Overall, this candidate vaccine did not elicit a durable humoral immune response.


Asunto(s)
Vacunas contra el Dengue/normas , Dengue/prevención & control , Esquemas de Inmunización , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/sangre , Niño , Preescolar , Dengue/epidemiología , Vacunas contra el Dengue/administración & dosificación , Vacunas contra el Dengue/inmunología , Humanos , Inmunización Secundaria , Lactante , Tailandia/epidemiología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/normas
11.
J Infect Dis ; 213(10): 1562-72, 2016 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-26704612

RESUMEN

BACKGROUND: A safe, effective tetravalent dengue vaccine is a global health priority. The safety and immunogenicity of a live attenuated, recombinant tetravalent dengue vaccine candidate (TDV) were evaluated in healthy volunteers from dengue-endemic countries. METHODS: This multicenter, double-blind, phase 2 study was conducted in Puerto Rico, Colombia, Singapore, and Thailand. During stage I, 148 volunteers aged 1.5-45 years were sequentially enrolled into 4 age-descending groups and randomized at a ratio of 2:1 to receive TDV or placebo. In stage II (group 5), 212 children aged 1.5-11 years were randomized at a ratio of 3:1 to receive TDV or placebo. Participants received a subcutaneous injection of TDV or placebo on days 0 and 90 and were followed for analysis of safety, seropositivity, and neutralizing antibodies to DENV-1-4. RESULTS: Injection site pain, itching, and erythema (mostly mild) were the only solicited adverse events more frequently reported with TDV than with placebo in all age groups. After 2 TDV doses, seropositivity was >95% in all 5 groups for DENV-1-3 and 72.7%-100% for DENV-4; geometric mean titers ranged from 582 to 1187 for DENV-1, from 582 to 1187 for DENV-2, from 196 to 630 for DENV-3, and from 41 to 210 for DENV-4 among the 5 groups. CONCLUSIONS: TDV was well tolerated and immunogenic in volunteers aged 1.5-45 years, irrespective of prevaccination dengue exposure.


Asunto(s)
Anticuerpos Antivirales/inmunología , Vacunas contra el Dengue/inmunología , Virus del Dengue/inmunología , Dengue/prevención & control , Adolescente , Adulto , Anticuerpos Neutralizantes/inmunología , Niño , Preescolar , Colombia , Dengue/inmunología , Vacunas contra el Dengue/administración & dosificación , Vacunas contra el Dengue/normas , Método Doble Ciego , Femenino , Humanos , Lactante , Inyecciones Subcutáneas , Masculino , Persona de Mediana Edad , Puerto Rico , Seguridad , Singapur , Tailandia , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/normas , Adulto Joven
12.
Vaccine ; 33(39): 5127-34, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26279339

RESUMEN

BACKGROUND: The recombinant yellow fever-17D-dengue virus, live, attenuated, tetravalent dengue vaccine (CYD-TDV) has undergone extensive clinical trials. Here safety and consistency of immunogenicity of phase III manufacturing lots of CYD-TDV were evaluated and compared with a phase II lot and placebo in a dengue-naïve population. METHODS: Healthy 18-60 year-olds were randomly assigned in a 3:3:3:3:1 ratio to receive three subcutaneous doses of either CYD-TDV from any one of three phase III lots or a phase II lot, or placebo, respectively in a 0, 6, 12 month dosing schedule. Neutralising antibody geometric mean titres (PRNT50 GMTs) for each of the four dengue serotypes were compared in sera collected 28 days after the third vaccination-equivalence among lots was demonstrated if the lower and upper limits of the two-sided 95% CIs of the GMT ratio were ≥0.5 and ≤2.0, respectively. RESULTS: 712 participants received vaccine or placebo and 614 (86%) completed the study; 17 (2.4%) participants withdrew after adverse events. Equivalence of phase III lots was demonstrated for 11 of 12 pairwise comparisons. One of three comparisons for serotype 2 was not statistically equivalent. GMTs for serotype 2 in phase III lots were close to each other (65.9, 44.1 and 58.1, respectively). CONCLUSIONS: Phase III lots can be produced in a consistent manner with predictable immune response and acceptable safety profile similar to previously characterised phase II lots. The phase III lots may be considered as not clinically different as statistical equivalence was shown for serotypes 1, 3 and 4 across the phase III lots. For serotype 2, although equivalence was not shown between two lots, the GMTs observed in the phase III lots were consistently higher than those for the phase II lot. As such, in our view, biological equivalence for all serotypes was demonstrated.


Asunto(s)
Vacunas contra el Dengue/efectos adversos , Vacunas contra el Dengue/inmunología , Dengue/prevención & control , Adolescente , Adulto , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Australia , Vacunas contra el Dengue/administración & dosificación , Vacunas contra el Dengue/normas , Femenino , Voluntarios Sanos , Humanos , Inyecciones Subcutáneas , Masculino , Persona de Mediana Edad , Placebos/administración & dosificación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/normas , Adulto Joven
13.
PLoS Negl Trop Dis ; 9(6): e0003749, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26065421

RESUMEN

Dengue is the most prevalent mosquito-borne viral disease worldwide. Yet, there are no vaccines or specific antivirals available to prevent or treat the disease. Several dengue vaccines are currently in clinical or preclinical stages. The most advanced vaccine is the chimeric tetravalent CYD-TDV vaccine of Sanofi Pasteur. This vaccine has recently cleared Phase III, and efficacy results have been published. Excellent tetravalent seroconversion was seen, yet the protective efficacy against infection was surprisingly low. Here, we will describe the complicating factors involved in the generation of a safe and efficacious dengue vaccine. Furthermore, we will discuss the human antibody responses during infection, including the epitopes targeted in humans. Also, we will discuss the current understanding of the assays used to evaluate antibody response. We hope this review will aid future dengue vaccine development as well as fundamental research related to the phenomenon of antibody-dependent enhancement of dengue virus infection.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Vacunas contra el Dengue/normas , Virus del Dengue/inmunología , Dengue/prevención & control , Acrecentamiento Dependiente de Anticuerpo , Dengue/inmunología , Vacunas contra el Dengue/inmunología , Virus del Dengue/fisiología , Epítopos/inmunología , Humanos , Linfocitos T/inmunología
14.
PLoS Negl Trop Dis ; 7(5): e2243, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23738026

RESUMEN

BACKGROUND: We have developed a manufacturing strategy that can improve the safety and genetic stability of recombinant live-attenuated chimeric dengue vaccine (DENVax) viruses. These viruses, containing the pre-membrane (prM) and envelope (E) genes of dengue serotypes 1-4 in the replicative background of the attenuated dengue-2 PDK-53 vaccine virus candidate, were manufactured under cGMP. METHODOLOGY/PRINCIPAL FINDINGS: After deriving vaccine viruses from RNA-transfected Vero cells, six plaque-purified viruses for each serotype were produced. The plaque-purified strains were then analyzed to select one stock for generation of the master seed. Full genetic and phenotypic characterizations of the master virus seeds were conducted to ensure these viruses retained the previously identified attenuating determinants and phenotypes of the vaccine viruses. We also assessed vector competence of the vaccine viruses in sympatric (Thai) Aedes aegypti mosquito vectors. CONCLUSION/SIGNIFICANCE: All four serotypes of master vaccine seeds retained the previously defined safety features, including all three major genetic loci of attenuation, small plaques, temperature sensitivity in mammalian cells, reduced replication in mosquito cell cultures, and reduced neurovirulence in new-born mice. In addition, the candidate vaccine viruses demonstrated greatly reduced infection and dissemination in Aedes aegypti mosquitoes, and are not likely to be transmissible by these mosquitoes. This manufacturing strategy has successfully been used to produce the candidate tetravalent vaccine, which is currently being tested in human clinical trials in the United States, Central and South America, and Asia.


Asunto(s)
Vacunas contra el Dengue/genética , Vacunas contra el Dengue/inmunología , Virus del Dengue/genética , Virus del Dengue/inmunología , Aedes , Animales , Animales Recién Nacidos , Línea Celular , Dengue/patología , Dengue/virología , Vacunas contra el Dengue/efectos adversos , Vacunas contra el Dengue/normas , Modelos Animales de Enfermedad , Femenino , Inestabilidad Genómica , Ratones , Ratones Endogámicos ICR , Control de Calidad , Tecnología Farmacéutica/métodos , Temperatura , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/normas , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/normas , Ensayo de Placa Viral , Virulencia , Replicación Viral/efectos de la radiación
15.
Comp Immunol Microbiol Infect Dis ; 31(2-3): 239-52, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17889365

RESUMEN

The increased incidence and extended geographical reach of Dengue virus over the past two decades have made the development of an effective vaccine an international urgency. Various strategies are being pursued, including live, vectored and killed/recombinant preparations. For all approaches, the challenge is to induce a broad durable immune response against all four serotypes of Dengue virus simultaneously whilst avoiding the possible exacerbation of risk of developing the severe forms of disease through incomplete or modified responses. This review presents the current state of knowledge and discusses the challenges of further clinical development.


Asunto(s)
Vacunas contra el Dengue/normas , Virus del Dengue/inmunología , Dengue/prevención & control , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales , Vacunas contra el Dengue/inmunología , Humanos , Inmunidad Celular , Modelos Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...