Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
BMC Med ; 18(1): 174, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32611419

RESUMO

BACKGROUND: Infection with cytomegalovirus (CMV) is highly prevalent worldwide and can cause severe disease in immunocompromised persons and congenitally infected infants. The disease burden caused by congenital CMV infection is high, especially in resource-limited countries. Vaccines are currently under development for various target groups. METHODS: We evaluated the impact of vaccination strategies and hygiene intervention using transmission models. Model parameters were estimated from a cross-sectional serological population study (n=5179) and a retrospective birth cohort (n=31,484), providing information on the age- and sex-specific CMV prevalence and on the birth prevalence of congenital CMV (cCMV). RESULTS: The analyses show that vertical transmission and infectious reactivation are the main drivers of transmission. Vaccination strategies aimed at reducing transmission from mother to child (vaccinating pregnant women or women of reproductive age) can yield substantial reductions of cCMV in 20 years (31.7-71.4% if 70% of women are effectively vaccinated). Alternatively, hygiene intervention aimed at preventing CMV infection and re-infection of women of reproductive age from young children is expected to reduce cCMV by less than 2%. The effects of large-scale vaccination on CMV prevalence can be substantial, owing to the moderate transmissibility of CMV at the population level. However, as CMV causes lifelong infection, the timescale on which reductions in CMV prevalence are expected is in the order of several decades. Elimination of CMV infection in the long run is only feasible for a vaccine with a long duration of protection and high vaccination coverage. CONCLUSIONS: Vaccination is an effective intervention to reduce the birth prevalence of cCMV. Population-level reductions in CMV prevalence can only be achieved on a long timescale. Our results stress the value of vaccinating pregnant women and women of childbearing age and provide support for the development of CMV vaccines and early planning of vaccination scenarios and rollouts.


Assuntos
Infecções por Citomegalovirus/transmissão , Vacinas contra Citomegalovirus/uso terapêutico , Citomegalovirus/imunologia , Estudos Transversais , Infecções por Citomegalovirus/virologia , Vacinas contra Citomegalovirus/farmacologia , Feminino , Humanos , Masculino , Estudos Retrospectivos
2.
Proc Natl Acad Sci U S A ; 116(26): 13036-13041, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31189602

RESUMO

Human cytomegalovirus (HCMV) causes severe disease in infants and immunocompromised people. There is no approved HCMV vaccine, and vaccine development strategies are complicated by evidence of both persistent infection and reinfection of people with prior immunity. The greatest emphasis has been placed on reducing transmission to seronegative pregnant women to prevent vertical transmission and its potentially severe sequelae. Increasing evidence suggests that the earliest host-HCMV interactions establish conditions for viral persistence, including evasion of host immune responses to the virus. Using a nonhuman primate model of HCMV infection, we show that rhesus macaques immunized against viral interleukin-10 (IL-10) manifest delayed rhesus cytomegalovirus (RhCMV) acquisition and altered immune responses to the infection when it does occur. Among animals with the greatest antiviral IL-10-neutralizing activity, the timing of RhCMV seroconversion was delayed by an average of 12 weeks. After acquisition, such animals displayed an antibody response to the new infection, which peaked as expected after 2 weeks but then declined rapidly. In contrast, surprisingly, vaccination with glycoprotein B (gB) protein had no discernible impact on these outcomes. Our results demonstrate that viral IL-10 is a key regulator of successful host immune responses to RhCMV. Viral IL-10 is, therefore, an important target for vaccine strategies against cytomegalovirus (CMV). Furthermore, given the immunoregulatory function of viral IL-10, targeting this protein may prove synergistic with other vaccine therapies and targets. Our study also provides additional evidence that the earliest host-CMV interactions can have a significant impact on the nature of persistent infection.


Assuntos
Antígenos Virais/imunologia , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/farmacologia , Citomegalovirus/imunologia , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Interleucina-10/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Antígenos Virais/administração & dosagem , Infecções por Citomegalovirus/sangue , Infecções por Citomegalovirus/transmissão , Infecções por Citomegalovirus/virologia , Vacinas contra Citomegalovirus/imunologia , Vacinas contra Citomegalovirus/uso terapêutico , Modelos Animais de Doenças , Feminino , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade nas Mucosas , Imunogenicidade da Vacina , Interleucina-10/administração & dosagem , Macaca mulatta , Gravidez , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Proteínas do Envelope Viral/administração & dosagem , Eliminação de Partículas Virais/imunologia
3.
Ital J Pediatr ; 44(1): 31, 2018 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-29490704

RESUMO

BACKGROUND: Since there is no effective treatment or vaccine against the congenital cytomegalovirus (cCMV) infection, knowledge and awareness of medical doctor's (MDs) especially family doctors are essential for preventive strategies and it also seems to be usually ignored by healthcare providers. Aim of this study was to investigate awareness of MDs about cCMV infection in Iran. METHODS: A single page questionnaire was randomly distributed among 450 MDs including general practitioners, pediatricians, gynecologists, internal and other medical specialists concerning of their knowledge in clinical presentation, diagnosis, prevention, prognosis, epidemiology, transmission, and management of cCMV infection. All statistical analyses were performed using SPSS version 16. RESULTS: More than half of questionnaire recipients refused to take part in any of the questionnaire items. The most of the respondents were agreed for newborn CMV screening tests and mandatory CMV test for women trying to get pregnant, which, are not routinely tested. The knowledge of general practitioners about cCMV was less than usual. The field of expertise had a profound effect in this survey, but age and gender did not. CONCLUSIONS: Our results indicated that the knowledge of cCMV infection, especially among family doctors contains several gaps. Urgent action is required to improve family doctor's knowledge of CMV infection. Surveys to evaluate CMV awareness among MDs, healthcare professionals and women of childbearing age are proposed.


Assuntos
Competência Clínica , Infecções por Citomegalovirus/congênito , Vacinas contra Citomegalovirus/farmacologia , Citomegalovirus/imunologia , Médicos de Família/normas , Inquéritos e Questionários , Adulto , Estudos Transversais , Infecções por Citomegalovirus/epidemiologia , Infecções por Citomegalovirus/prevenção & controle , Feminino , Humanos , Incidência , Irã (Geográfico)/epidemiologia , Masculino
4.
Expert Opin Drug Metab Toxicol ; 7(10): 1245-65, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21883024

RESUMO

INTRODUCTION: Cytomegalovirus (CMV) is a ubiquitous pathogen that establishes a lifelong asymptomatic infection in healthy individuals. Infection of immunesuppressed individuals causes serious illness. Transplant and AIDS patients are highly susceptible to CMV leading to life-threatening end-organ disease. Another vulnerable population is the developing fetus in utero, where congenital infection can result in surviving newborns with long-term developmental problems. There is no vaccine licensed for CMV and current antivirals suffer from complications associated with prolonged treatment. These include drug toxicity and emergence of resistant strains. There is an obvious need for new antivirals. Candidate intervention strategies are tested in controlled preclinical animal models but species specificity of human CMV precludes the direct study of the virus in an animal model. AREAS COVERED: This review explores the current status of CMV antivirals and development of new drugs. This includes the use of animal models and the development of new improved models such as humanized animal CMV and bioluminescent imaging of virus in animals in real time. EXPERT OPINION: Various new CMV antivirals are in development, some with greater spectrum of activity against other viruses. Although the greatest need is in the setting of transplant patients, there remains an unmet need for a safe antiviral strategy against congenital CMV. This is especially important as an effective CMV vaccine remains an elusive goal. In this regard, greater emphasis should be placed on suitable preclinical animal models and greater collaboration between industry and academia.


Assuntos
Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Animais , Antivirais/efeitos adversos , Citomegalovirus/isolamento & purificação , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/farmacologia , Avaliação Pré-Clínica de Medicamentos , Cobaias , Humanos , Modelos Animais
5.
Lancet ; 377(9773): 1256-63, 2011 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-21481708

RESUMO

BACKGROUND: Cytomegalovirus end-organ disease can be prevented by giving ganciclovir when viraemia is detected in allograft recipients. Values of viral load correlate with development of end-organ disease and are moderated by pre-existing natural immunity. Our aim was to determine whether vaccine-induced immunity could do likewise. METHODS: We undertook a phase-2 randomised placebo controlled trial in adults awaiting kidney or liver transplantation at the Royal Free Hospital, London, UK. Exclusion criteria were pregnancy, receipt of blood products (except albumin) in the previous 3 months, and simultaneous multiorgan transplantation. 70 patients seronegative and 70 seropositive for cytomegalovirus were randomly assigned from a scratch-off randomisation code in a 1:1 ratio to receive either cytomegalovirus glycoprotein-B vaccine with MF59 adjuvant or placebo, each given at baseline, 1 month and 6 months later. If a patient was transplanted, no further vaccinations were given and serial blood samples were tested for cytomegalovirus DNA by real-time quantitative PCR (rtqPCR). Any patient with one blood sample containing more than 3000 cytomegalovirus genomes per mL received ganciclovir until two consecutive undetectable cytomegalovirus DNA measurements. Safety and immunogenicity were coprimary endpoints and were assessed by intention to treat in patients who received at least one dose of vaccine or placebo. This trial is registered with ClinicalTrials.gov, NCT00299260. FINDINGS: 67 patients received vaccine and 73 placebo, all of whom were evaluable. Glycoprotein-B antibody titres were significantly increased in both seronegative (geometric mean titre 12,537 (95% CI 6593-23,840) versus 86 (63-118) in recipients of placebo recipients; p<0.0001) and seropositive (118,395; 64,503-217,272) versus 24,682 (17,909-34,017); p<0.0001) recipients of vaccine. In those who developed viraemia after transplantation, glycoprotein-B antibody titres correlated inversely with duration of viraemia (p=0.0022). In the seronegative patients with seropositive donors, the duration of viraemia (p=0.0480) and number of days of ganciclovir treatment (p=0.0287) were reduced in vaccine recipients. INTERPRETATION: Although cytomegalovirus disease occurs in the context of suppressed cell-mediated immunity post-transplantation, humoral immunity has a role in reduction of cytomegalovirus viraemia. Vaccines containing cytomegalovirus glycoprotein B merit further assessment in transplant recipients. FUNDING: National Institute of Allergy and Infectious Diseases, Grant R01AI051355 and Wellcome Trust, Grant 078332. SPONSOR: University College London (UCL).


Assuntos
Infecções por Citomegalovirus/diagnóstico , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/administração & dosagem , Citomegalovirus/isolamento & purificação , Imunossupressores/efeitos adversos , Transplante de Órgãos , Polissorbatos/administração & dosagem , Esqualeno/administração & dosagem , Proteínas do Envelope Viral/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Adulto , Idoso , Anticorpos Antivirais/isolamento & purificação , Citomegalovirus/genética , Citomegalovirus/imunologia , Vacinas contra Citomegalovirus/farmacologia , DNA Viral/isolamento & purificação , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imunossupressores/administração & dosagem , Transplante de Rim , Transplante de Fígado , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Fatores de Tempo , Resultado do Tratamento , Proteínas do Envelope Viral/farmacologia , Viremia/diagnóstico , Viremia/prevenção & controle
6.
PLoS One ; 6(1): e15949, 2011 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-21264339

RESUMO

BACKGROUND: Infection with multiple CMV strains is common in immunocompromised hosts, but its occurrence in normal hosts has not been well-studied. METHODS: We analyzed CMV strains longitudinally in women who acquired CMV while enrolled in a CMV glycoprotein B (gB) vaccine trial. Sequencing of four variable genes was performed in samples collected from seroconversion and up to 34 months thereafter. RESULTS: 199 cultured isolates from 53 women and 65 original fluids from a subset of 19 women were sequenced. 51 women were infected with one strain each without evidence for genetic drift; only two women shed multiple strains. Genetic variability among strains increased with the number of sequenced genetic loci. Nevertheless, 13 of 53 women proved to be infected with an identical CMV strain based on sequencing at all four variable genes. CMV vaccine did not alter the degree of genetic diversity amongst strains. CONCLUSIONS: Primary CMV infection in healthy women nearly always involves shedding of one strain that remains stable over time. Immunization with CMVgB-1 vaccine strain is not selective against specific strains. Although 75% of women harbored their unique strain, or a strain shared with only one other woman, 25% shared a single common strain, suggesting that this predominant strain with a particular combination of genetic loci is advantageous in this large urban area.


Assuntos
Citomegalovirus/genética , Citomegalovirus/isolamento & purificação , Variação Genética , Adolescente , Adulto , Sequência de Bases , Infecções por Citomegalovirus/virologia , Vacinas contra Citomegalovirus/farmacologia , Feminino , Humanos , Estudos Longitudinais , Dados de Sequência Molecular , Análise de Sequência de DNA , Especificidade da Espécie , Proteínas Virais/imunologia , Proteínas Virais/uso terapêutico , Adulto Jovem
7.
Vaccine ; 24(37-39): 6175-86, 2006 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-16879902

RESUMO

It is unclear if protective immunity can be conferred by a cytomegalovirus (CMV) vaccine encoding a single protein subunit, or if multiple viral genes need to be targeted. Using the guinea pig model of congenital CMV infection, these studies examined the immunogenicity and efficacy of a DNA vaccine based on the guinea pig cytomegalovirus (GPCMV) genome cloned as a non-infectious BAC plasmid, modified by transposon insertion into the homolog of the HCMV tegument protein, UL48. Following vaccination of female Hartley guinea pigs with BAC DNA, adverse GPCMV-related pregnancy outcome were assessed after establishment of pregnancy, followed by GPCMV third-trimester challenge. Animals immunized with recombinant BACmid engendered anti-GPCMV antibodies by ELISA assay. Immunogenicity of BAC plasmid DNA was augmented by inclusion of the lipid adjuvant, DOTMA/DOPE, in the vaccine regimen. Among pups born to 12 control (sham-immunized) dams challenged with GPCMV in the third trimester, mortality was 23/35 (66%). In contrast, among evaluable pregnancy outcomes in pups born to 10 BAC-immunized pregnant dams, preconception immunization resulted in reduced pup mortality, to 10/34 pups (29%; p<0.005 versus control, Fisher's exact test). In addition, vaccinated dams had reduced viral load, compared to controls, as assessed by quantitative, real-time PCR.


Assuntos
Infecções por Citomegalovirus/congênito , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/imunologia , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Complicações Infecciosas na Gravidez/imunologia , Roseolovirus/imunologia , Vacinas de DNA/imunologia , Sequência de Aminoácidos , Animais , Cromossomos Artificiais Bacterianos/genética , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/transmissão , Vacinas contra Citomegalovirus/genética , Vacinas contra Citomegalovirus/farmacologia , Elementos de DNA Transponíveis/genética , Elementos de DNA Transponíveis/imunologia , Escherichia coli/genética , Feminino , Genoma Viral/imunologia , Cobaias , Camundongos , Dados de Sequência Molecular , Gravidez , Complicações Infecciosas na Gravidez/virologia , Roseolovirus/genética , Vacinas de DNA/genética , Vacinas de DNA/farmacologia , Carga Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA