Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vaccine ; 36(22): 3090-3100, 2018 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-28216187

RESUMO

Infections with filoviruses in humans are highly virulent, causing hemorrhagic fevers which result in up to 90% mortality. In addition to natural infections, the ability to use these viruses as bioterrorist weapons is of significant concern. Currently, there are no licensed vaccines or therapeutics available to combat these infections. The pathogenesis of disease involves the dysregulation of the host's immune system, which results in impairment of the innate and adaptive immune responses, with subsequent development of lymphopenia, thrombocytopenia, hemorrhage, and death. Questions remain with regard to the few survivors of infection, who manage to mount an effective adaptive immune response. These questions concern the humoral and cellular components of this response, and whether such a response can be elicited by an appropriate prophylactic vaccine. The data reported herein describe the production and evaluation of a recombinant subunit Ebola virus vaccine candidate consisting of insect cell expressed Zaire ebolavirus (EBOV) surface glycoprotein (GP) and the matrix proteins VP24 and VP40. The recombinant subunit proteins are shown to be highly immunogenic in mice, yielding both humoral and cellular responses, as well as highly efficacious, providing up to 100% protection against a lethal challenge with live virus. These results demonstrate proof of concept for such a recombinant non-replicating vaccine candidate in the mouse model of EBOV which helps to elucidate immune correlates of protection and warrants further development.


Assuntos
Vacinas contra Ebola/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Proteínas do Envelope Viral/imunologia , Proteínas da Matriz Viral/imunologia , Proteínas Virais/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Ebolavirus , Feminino , Doença pelo Vírus Ebola/imunologia , Imunidade Celular , Imunidade Humoral , Camundongos , Camundongos Endogâmicos BALB C , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/imunologia
2.
Clin Vaccine Immunol ; 22(9): 1070-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26224691

RESUMO

The efficacy of a recombinant plague vaccine (rF1V) was evaluated in cynomolgus macaques (CMs) to establish the relationship among vaccine doses, antibody titers, and survival following an aerosol challenge with a lethal dose of Yersinia pestis strain Colorado 92. CMs were vaccinated with a range of rF1V doses on a three-dose schedule (days 0, 56, and 121) to provide a range of survival outcomes. The humoral immune response following vaccination was evaluated with anti-rF1, anti-rV, and anti-rF1V bridge enzyme-linked immunosorbent assays (ELISAs). Animals were challenged via aerosol exposure on day 149. Vaccine doses and antibody responses were each significantly associated with the probability of CM survival (P < 0.0001). Vaccination also decreased signs of pneumonic plague in a dose-dependent manner. There were statistically significant correlations between the vaccine dose and the time to onset of fever (P < 0.0001), the time from onset of fever to death (P < 0.0001), the time to onset of elevated respiratory rate (P = 0.0003), and the time to onset of decreased activity (P = 0.0251) postinfection in animals exhibiting these clinical signs. Delays in the onset of these clinical signs of disease were associated with larger doses of rF1V. Immunization with ≥ 12 µg of rF1V resulted in 100% CM survival. Since both the vaccine dose and anti-rF1V antibody titers correlate with survival, rF1V bridge ELISA titers can be used as a correlate of protection.


Assuntos
Anticorpos Antibacterianos/sangue , Vacina contra a Peste/administração & dosagem , Vacina contra a Peste/imunologia , Peste/imunologia , Peste/prevenção & controle , Yersinia pestis/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Imunidade Humoral , Esquemas de Imunização , Macaca fascicularis , Peste/microbiologia , Vacina contra a Peste/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
3.
Clin Vaccine Immunol ; 22(1): 46-55, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25355797

RESUMO

Studies on candidate pandemic vaccines against avian influenza viruses have focused on H5N1, but viruses of other subtypes, such as A/H9N2, are also considered to have pandemic potential. We investigated the safety and immunogenicity of two immunizations with one of five different antigen doses (ranging from 3.75 to 45 µg of hemagglutinin antigen) of a nonadjuvanted whole-virus G9 lineage H9N2 influenza virus vaccine in healthy adults aged 18 to 49 years. The antibody responses were measured by hemagglutination inhibition (HI), microneutralization (MN), and single radial hemolysis (SRH) assays. To investigate a hypothesis that previous exposure to H2N2 viruses in subjects born in or before 1968 might prime for more robust antibody responses to H9N2 vaccination than that in subjects born after 1968, a post hoc age-stratified analysis of antibody responses was done. Both vaccinations in all dose groups were safe and well tolerated. No vaccine-related serious adverse events were reported, and the majority of the adverse reactions were rated as mild. The rates of injection site reactions were lower in the 3.75-µg- and 7.5-µg-dose groups than those in the higher-dose groups; the rates of systemic reactions were similar across all dose groups. The seroprotection rates among the different dose groups 21 days after the second immunization ranged from 52.8% to 88.9% as measured by HI assay, from 88.7% to 98.1% or 82.7% to 96.2% as measured by MN assay (MN titer cutoffs, 1:40 and 1:80, respectively), and from 94.2% to 100% as measured by SRH assay. Higher antibody responses were not induced in subjects born in or before 1968. These data indicate that a nonadjuvanted whole-virus H9N2 vaccine is well tolerated and immunogenic in healthy adults. (This study has been registered at ClinicalTrials.gov under registration no. NCT01320696.).


Assuntos
Vírus da Influenza A Subtipo H9N2/imunologia , Vacinas contra Influenza/efeitos adversos , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Vacinação/efeitos adversos , Vacinação/métodos , Adolescente , Adulto , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Método Duplo-Cego , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Feminino , Voluntários Saudáveis , Testes de Inibição da Hemaglutinação , Humanos , Imunodifusão , Vacinas contra Influenza/administração & dosagem , Masculino , Pessoa de Meia-Idade , Testes de Neutralização , Resultado do Tratamento , Adulto Jovem
4.
J Virol ; 88(20): 12077-86, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25122801

RESUMO

Alphavirus replicons were evaluated as potential vaccine candidates for Venezuelan equine encephalitis virus (VEEV), western equine encephalitis virus (WEEV), or eastern equine encephalitis virus (EEEV) when given individually or in combination (V/W/E) to mice or cynomolgus macaques. Individual replicon vaccines or the combination V/W/E replicon vaccine elicited strong neutralizing antibodies in mice to their respective alphavirus. Protection from either subcutaneous or aerosol challenge with VEEV, WEEV, or EEEV was demonstrated out to 12 months after vaccination in mice. Individual replicon vaccines or the combination V/W/E replicon vaccine elicited strong neutralizing antibodies in macaques and demonstrated good protection against aerosol challenge with an epizootic VEEV-IAB virus, Trinidad donkey. Similarly, the EEEV replicon and V/W/E combination vaccine elicited neutralizing antibodies against EEEV and protected against aerosol exposure to a North American variety of EEEV. Both the WEEV replicon and combination V/W/E vaccination, however, elicited poor neutralizing antibodies to WEEV in macaques, and the protection conferred was not as strong. These results demonstrate that a combination V/W/E vaccine is possible for protection against aerosol challenge and that cross-interference between the vaccines is minimal. Importance: Three related viruses belonging to the genus Alphavirus cause severe encephalitis in humans: Venezuelan equine encephalitis virus (VEEV), western equine encephalitis virus (WEEV), and eastern equine encephalitis virus (EEEV). Normally transmitted by mosquitoes, these viruses can cause disease when inhaled, so there is concern that these viruses could be used as biological weapons. Prior reports have suggested that vaccines for these three viruses might interfere with one another. We have developed a combined vaccine for Venezuelan equine encephalitis, western equine encephalitis, and eastern equine encephalitis expressing the surface proteins of all three viruses. In this report we demonstrate in both mice and macaques that this combined vaccine is safe, generates a strong immune response, and protects against aerosol challenge with the viruses that cause Venezuelan equine encephalitis, western equine encephalitis, and eastern equine encephalitis.


Assuntos
Alphavirus/imunologia , Anticorpos Neutralizantes/imunologia , Vírus da Encefalite Equina do Leste/imunologia , Replicon , Vacinas Virais/imunologia , Alphavirus/classificação , Animais , Western Blotting , Chlorocebus aethiops , Cricetinae , Vírus da Encefalite Equina do Leste/classificação , Ensaio de Imunoadsorção Enzimática , Feminino , Imunofluorescência , Macaca fascicularis , Masculino , Camundongos , Células Vero
5.
J Virol ; 87(9): 4952-64, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23408633

RESUMO

There are no vaccines or therapeutics currently approved for the prevention or treatment of ebolavirus infection. Previously, a replicon vaccine based on Venezuelan equine encephalitis virus (VEEV) demonstrated protective efficacy against Marburg virus in nonhuman primates. Here, we report the protective efficacy of Sudan virus (SUDV)- and Ebola virus (EBOV)-specific VEEV replicon particle (VRP) vaccines in nonhuman primates. VRP vaccines were developed to express the glycoprotein (GP) of either SUDV or EBOV. A single intramuscular vaccination of cynomolgus macaques with VRP expressing SUDV GP provided complete protection against intramuscular challenge with SUDV. Vaccination against SUDV and subsequent survival of SUDV challenge did not fully protect cynomolgus macaques against intramuscular EBOV back-challenge. However, a single simultaneous intramuscular vaccination with VRP expressing SUDV GP combined with VRP expressing EBOV GP did provide complete protection against intramuscular challenge with either SUDV or EBOV in cynomolgus macaques. Finally, intramuscular vaccination with VRP expressing SUDV GP completely protected cynomolgus macaques when challenged with aerosolized SUDV, although complete protection against aerosol challenge required two vaccinations with this vaccine.


Assuntos
Ebolavirus/imunologia , Vírus da Encefalite Equina Venezuelana/genética , Doença pelo Vírus Ebola/prevenção & controle , Replicon , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/imunologia , Ebolavirus/genética , Vírus da Encefalite Equina Venezuelana/fisiologia , Vetores Genéticos/genética , Vetores Genéticos/fisiologia , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Humanos , Macaca fascicularis , Vacinação , Proteínas do Envelope Viral/administração & dosagem , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
6.
PLoS One ; 7(10): e44769, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23056184

RESUMO

We report the rational design and in vivo testing of mosaic proteins for a polyvalent pan-filoviral vaccine using a computational strategy designed for the Human Immunodeficiency Virus type 1 (HIV-1) but also appropriate for Hepatitis C virus (HCV) and potentially other diverse viruses. Mosaics are sets of artificial recombinant proteins that are based on natural proteins. The recombinants are computationally selected using a genetic algorithm to optimize the coverage of potential cytotoxic T lymphocyte (CTL) epitopes. Because evolutionary history differs markedly between HIV-1 and filoviruses, we devised an adapted computational technique that is effective for sparsely sampled taxa; our first significant result is that the mosaic technique is effective in creating high-quality mosaic filovirus proteins. The resulting coverage of potential epitopes across filovirus species is superior to coverage by any natural variants, including current vaccine strains with demonstrated cross-reactivity. The mosaic cocktails are also robust: mosaics substantially outperformed natural strains when computationally tested against poorly sampled species and more variable genes. Furthermore, in a computational comparison of cross-reactive potential a design constructed prior to the Bundibugyo outbreak performed nearly as well against all species as an updated design that included Bundibugyo. These points suggest that the mosaic designs would be more resilient than natural-variant vaccines against future Ebola outbreaks dominated by novel viral variants. We demonstrate in vivo immunogenicity and protection against a heterologous challenge in a mouse model. This design work delineates the likely requirements and limitations on broadly-protective filoviral CTL vaccines.


Assuntos
Epitopos de Linfócito T/imunologia , Infecções por Filoviridae/imunologia , Filoviridae/imunologia , Linfócitos T Citotóxicos/imunologia , Vacinas Virais/imunologia , Vacinas contra a AIDS/imunologia , Animais , Anticorpos Antivirais/imunologia , Biologia Computacional/métodos , Reações Cruzadas/imunologia , Desenho de Fármacos , Vacinas contra Ebola/administração & dosagem , Vacinas contra Ebola/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Filoviridae/metabolismo , Infecções por Filoviridae/prevenção & controle , Infecções por Filoviridae/virologia , Hepacivirus/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sobrevida , Vacinas contra Hepatite Viral/imunologia , Proteínas Virais/imunologia , Vacinas Virais/administração & dosagem
7.
Clin Infect Dis ; 54(7): 946-54, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22267715

RESUMO

BACKGROUND: Current knowledge of the consistency of protection induced by seasonal influenza vaccines over the duration of a full influenza season is limited, and little is known about the clinical course of disease in individuals who become infected despite vaccination. METHODS: Data from a randomized double-blind placebo-controlled clinical trial undertaken in healthy young adults in the 2008-2009 influenza season were used to investigate the weekly cumulative efficacy of a Vero cell culture-derived influenza vaccine. In addition, the duration and severity of disease in vaccine and placebo recipients with cell culture-confirmed influenza infection were compared. RESULTS: Vaccine efficacy against matching strains was consistently high (73%-82%) throughout the study, including the entire period of the influenza season during which influenza activity was above the epidemic threshold. Vaccine efficacy was also consistent (68%-83%) when calculated for all strains, irrespective of antigenic match. Vaccination also ameliorated disease symptoms when infection was not prevented. Bivariate analysis of duration and severity showed a significant amelioration of myalgia (P = .003), headache (P = .025), and fatigue (P = .013) in infected vaccinated subjects compared with placebo. Cough (P = .143) and oropharyngeal pain (P = .083) were also reduced in infected vaccinated subjects. CONCLUSIONS: A Vero cell culture-derived influenza vaccine provides consistently high levels of protection against cell culture-confirmed infection by seasonal influenza virus and significantly reduces the duration and severity of disease in those individuals in which infection is not prevented. CLINICAL TRIALS REGISTRATION: ClinicalTrials.gov NCT00566345.


Assuntos
Vacinas contra Influenza/imunologia , Influenza Humana/patologia , Influenza Humana/prevenção & controle , Adulto , Animais , Biotecnologia/métodos , Técnicas de Cultura de Células/métodos , Chlorocebus aethiops , Método Duplo-Cego , Humanos , Vacinas contra Influenza/administração & dosagem , Pessoa de Meia-Idade , Placebos/administração & dosagem , Índice de Gravidade de Doença , Tecnologia Farmacêutica/métodos , Fatores de Tempo , Células Vero , Adulto Jovem
8.
Adv Prev Med ; 2012: 731604, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22028978

RESUMO

The development of vaccines for microorganisms and bacterial toxins with the potential to be used as biowarfare and bioterrorism agents is an important component of the US biodefense program. DVC is developing two vaccines, one against inhalational exposure to botulinum neurotoxins A1 and B1 and a second for Yersinia pestis, with the ultimate goal of licensure by the FDA under the Animal Rule. Progress has been made in all technical areas, including manufacturing, nonclinical, and clinical development and testing of the vaccines, and in assay development. The current status of development of these vaccines, and remaining challenges are described in this chapter.

9.
Vaccine ; 30(29): 4377-86, 2012 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-22172502

RESUMO

BACKGROUND: Cell culture technologies have the potential to improve the robustness and flexibility of influenza vaccine supply and to substantially shorten manufacturing timelines. We investigated the safety, immunogenicity and efficacy of a Vero cell culture-derived seasonal influenza vaccine and utilized these studies to establish a serological correlate of vaccine protection. METHODS: Two multicenter, randomized, double-blind phase III trials were undertaken in the US during the 2008-2009 Northern hemisphere influenza season, in young (18-49 years) and older (50-64 years and ≥ 65 years) adult subjects. 7250 young adults were randomized 1:1 to receive either Vero-derived vaccine or placebo. 3210 older adult subjects were randomized 8:1 to receive either Vero-derived vaccine or a licensed egg-derived vaccine. Serum hemagglutination inhibition antibody titers were assessed 21 days post-vaccination. Vaccine efficacy in preventing cell culture-confirmed influenza infection was determined for the young adult population. Local and systemic adverse events were recorded in both studies. RESULTS: The Vero-derived vaccine was safe and well tolerated in both young and older adults. All US and European immunological licensing thresholds were comfortably met in both populations. Vaccine efficacy in young adults was 79% against A/H1N1 viruses antigenically matching the corresponding vaccine strain and 78.5% for all antigenically matched influenza viruses. A hemagglutination inhibition antibody titer of ≥ 1:15 provided a reliable correlate of protection for the Vero-derived influenza vaccine, with no additional benefit at titers >1:30. Bridging of the correlate of protection established in the young adult population to the older adult immunogenicity data demonstrated the likely effectiveness of the Vero-derived vaccine in the older adult population. CONCLUSIONS: A Vero cell culture-derived seasonal influenza vaccine is safe, immunogenic and protects against infection with influenza virus. The novel vaccine technology has the potential to make a substantial contribution to improving influenza vaccine supply. CLINICAL TRIAL REGISTRATION: The studies are registered with ClinicalTrials.gov, numbers NCT00566345 and NCT00782431.


Assuntos
Vacinas contra Influenza/biossíntese , Influenza Humana/prevenção & controle , Adolescente , Adulto , Idoso , Animais , Anticorpos Antivirais/sangue , Técnicas de Cultura de Células , Chlorocebus aethiops , Método Duplo-Cego , Feminino , Testes de Inibição da Hemaglutinação , Humanos , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/efeitos adversos , Masculino , Pessoa de Meia-Idade , Células Vero , Adulto Jovem
10.
Lancet ; 377(9767): 751-9, 2011 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-21329971

RESUMO

BACKGROUND: The use of cell-culture technologies for the manufacture of influenza vaccines might contribute to improved strain selection and robust vaccine supplies. We investigated the safety, immunogenicity, and protective efficacy of a Vero-cell-culture-derived influenza vaccine, and assessed the correlation between vaccine efficacy and haemagglutination inhibition antibody titre. METHODS: In a double-blind, placebo-controlled, phase 3 trial undertaken in 36 centres in the USA, healthy adults (aged 18-49 years) were randomly assigned in a 1:1 ratio to one injection of either placebo or Vero-cell-culture-derived influenza vaccine during the 2008-09 season. Randomisation was done in blocks by use of the random number generator algorithm, and participants were allocated by use of a centralised telephone system. The primary objective was the efficacy of the vaccine in preventing cell-culture-confirmed influenza infection with viruses that were antigenically matched to one of the vaccine strains. Analysis was by intention to treat. The study is registered with ClinicalTrials.gov, number NCT00566345. FINDINGS: 7250 participants were randomly assigned to vaccine (n=3626) and placebo (n=3624). 7236 were analysed for the primary outcome (n=3619 and n=3617, respectively). Overall protective efficacy for antigenically matched influenza infection was 78·5% (95% CI 60·8-88·2). The vaccine was well tolerated with no treatment-related serious adverse events. Adverse events were mainly mild and transient. An HI titre of at least 1:15 provided a reliable correlate of cell-culture-derived influenza vaccine-induced protection; no additional benefit was noted with titres greater than 1:30. INTERPRETATION: The data indicate that existing correlates of protection afforded with egg-derived seasonal influenza vaccines also apply to this vaccine. FUNDING: Federal (US Government) funds from the Office for Preparedness and Response, Biomedical Advanced Research and Development Authority, under contract to DynPort Vaccine Company.


Assuntos
Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Células Vero , Adulto , Animais , Chlorocebus aethiops , Método Duplo-Cego , Feminino , Testes de Inibição da Hemaglutinação , Humanos , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/efeitos adversos , Influenza Humana/induzido quimicamente , Influenza Humana/imunologia , Masculino , Pessoa de Meia-Idade , Curva ROC , Medição de Risco , Sensibilidade e Especificidade , Resultado do Tratamento , Estados Unidos
11.
J Virol ; 84(24): 12683-90, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20926570

RESUMO

Six monoclonal antibodies were isolated that exhibited specificity for a furin cleavage site deletion mutant (V3526) of Venezuelan equine encephalitis virus (VEEV). These antibodies comprise a single competition group and bound the E3 glycoprotein of VEEV subtype I viruses but failed to bind the E3 glycoprotein of other alphaviruses. These antibodies neutralized V3526 virus infectivity but did not neutralize the parental strain of Trinidad donkey (TrD) VEEV. However, the E3-specific antibodies did inhibit the production of virus from VEEV TrD-infected cells. In addition, passive immunization of mice demonstrated that antibody to the E3 glycoprotein provided protection against lethal VEEV TrD challenge. This is the first recognition of a protective epitope in the E3 glycoprotein. Furthermore, these results indicate that E3 plays a critical role late in the morphogenesis of progeny virus after E3 appears on the surfaces of infected cells.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Antivirais/administração & dosagem , Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/prevenção & controle , Glicoproteínas/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Ligação Competitiva , Encefalomielite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/virologia , Epitopos/imunologia , Técnica Indireta de Fluorescência para Anticorpo , Glicoproteínas/antagonistas & inibidores , Imunização Passiva , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Envelope Viral/antagonistas & inibidores
12.
Vaccine ; 28(49): 7748-56, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-20920572

RESUMO

Passive transfer models were developed to evaluate the ability of antibodies generated in cynomolgus macaques and humans vaccinated with a recombinant plague vaccine (rF1V) to protect naïve Swiss Webster mice against pneumonic plague. Development of the passive transfer model is intended to support clinical and nonclinical development of the rF1V vaccine. To evaluate protection, unfractionated serum collected from rF1V vaccinated cynomolgus macaques and human volunteers with known antibody titers to rF1, rV and rF1V was transferred into naïve Swiss Webster mice via the intraperitoneal route. Results of these studies demonstrated that passive immunization protected mice from challenge or extended mean survival time and that the passive transfer assay can be used to evaluate the functional role of antibodies induced by rF1V vaccination in protection against aerosol exposure.


Assuntos
Anticorpos Antibacterianos/imunologia , Imunização Passiva , Vacina contra a Peste/imunologia , Peste/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Ensaio de Imunoadsorção Enzimática , Humanos , Imunidade Humoral , Macaca fascicularis , Camundongos , Peste/imunologia , Análise de Sobrevida , Vacinas Sintéticas/imunologia , Yersinia pestis/imunologia
13.
Vaccine ; 28(18): 3143-51, 2010 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-20193792

RESUMO

V3526, a genetically modified strain of Venezuelan equine encephalitis virus (VEEV), was formalin inactivated for evaluation as a next generation vaccine candidate for VEEV. In this study, we tested formalin-inactivated V3526 (fV3526) with and without adjuvant for immunogenicity and efficacy in BALB/c mice and results were compared to the existing inactivated VEEV vaccine, C84. Mice were vaccinated intramuscularly (IM) or subcutaneously (SC) with fV3526 formulations and challenged with VEEV IAB Trinidad donkey (VEEV TrD) strain by SC or aerosol exposure. Efficacy following SC or aerosol challenge was not significantly different between the fV3526 formulations or compared to C84 despite C84 being administered in more doses and higher concentration of viral protein per dose. These data support further evaluation of fV3526 formulations as a next generation VEEV vaccine.


Assuntos
Vírus da Encefalite Equina Venezuelana/imunologia , Vacinas Virais/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Desinfetantes/farmacologia , Vírus da Encefalite Equina Venezuelana/genética , Encefalomielite Equina Venezuelana/prevenção & controle , Feminino , Formaldeído/farmacologia , Injeções Intramusculares , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sobrevida , Vacinas de Produtos Inativados/imunologia , Vacinas Virais/administração & dosagem
14.
Vaccine ; 28(4): 1031-40, 2010 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-19914193

RESUMO

We recently developed a gamma-irradiation method to inactivate V3526, a live-attenuated Venezuelan equine encephalitis virus (VEEV) vaccine candidate. Dosage and schedule studies were conducted to evaluate the immunogenicity and efficacy of gamma-irradiated V3526 (gV3526). Subcutaneous (SC) and low dosage intramuscular (IM) administration of gV3526 were highly effective in protecting mice against a SC challenge with VEEV IA/B Trinidad Donkey strain, but not against an equivalent aerosol challenge. More robust immune responses and increased protective efficacy were noted when the IM dosage of gV3526 was increased. IM administration of gV3526 formulated with either CpG or CpG plus Alhydrogel further augmented the immune response in mice and resulted in 100% protection against aerosol challenge.


Assuntos
Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/prevenção & controle , Vacinas Virais/imunologia , Adjuvantes Imunológicos/administração & dosagem , Hidróxido de Alumínio/administração & dosagem , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Relação Dose-Resposta Imunológica , Vírus da Encefalite Equina Venezuelana/efeitos da radiação , Raios gama , Injeções Intramusculares , Injeções Subcutâneas , Camundongos , Oligodesoxirribonucleotídeos/administração & dosagem , Análise de Sobrevida , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Vacinas Virais/administração & dosagem , Inativação de Vírus
15.
Vaccine ; 27(49): 6814-23, 2009 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-19761841

RESUMO

Non-human primates (NHP) are considered to be the most appropriate model for predicting how humans will respond to many infectious diseases. Due to ethical and monetary concerns associated with the use of NHP, rodent models that are as predictive of responses likely to be seen in human vaccine recipients are warranted. Using implanted telemetry devices, body temperature and activity were monitored in inbred and outbred mouse strains following administration of the live-attenuated vaccine for Venezuelan equine encephalitis virus (VEEV), V3526. Following analysis of individual mouse data, only outbred mouse strains showed changes in diurnal temperature and activity profiles following vaccination. Similar changes were observed following VEEV challenge of vaccinated outbred mice. From these studies, we conclude, outbred mouse strains implanted with telemeters are a sensitive model for predicting responses in humans following vaccination.


Assuntos
Temperatura Corporal , Febre/imunologia , Telemetria , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Comportamento Animal , Peso Corporal , Modelos Animais de Doenças , Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/prevenção & controle , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Sensibilidade e Especificidade , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Vacinas Virais/administração & dosagem
16.
J Mol Biol ; 375(1): 202-16, 2008 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-18005986

RESUMO

13F6-1-2 is a murine monoclonal antibody that recognizes the heavily glycosylated mucin-like domain of the Ebola virus virion-attached glycoprotein (GP) and protects animals against lethal viral challenge. Here we present the crystal structure, at 2.0 A, of 13F6-1-2 in complex with its Ebola virus GP peptide epitope. The GP peptide binds in an extended conformation, anchored primarily by interactions with the heavy chain. Two GP residues, Gln P406 and Arg P409, make extensive side-chain hydrogen bond and electrostatic interactions with the antibody and are likely critical for recognition and affinity. The 13F6-1-2 antibody utilizes a rare V lambda(x) light chain. The three light-chain complementarity-determining regions do not adopt canonical conformations and represent new classes of structures distinct from V kappa and other V lambda light chains. In addition, although V lambda(x) had been thought to confer specificity, all light-chain contacts are mediated through germ-line-encoded residues. This structure of an antibody that protects against the Ebola virus now provides a framework for humanization and development of a postexposure immunotherapeutic.


Assuntos
Anticorpos Monoclonais/imunologia , Ebolavirus/química , Epitopos , Glicoproteínas/imunologia , Cadeias lambda de Imunoglobulina/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Arginina/metabolismo , Sequência de Bases , Códon de Terminação , Dissulfetos/química , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Ligação de Hidrogênio , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/metabolismo , Cadeias lambda de Imunoglobulina/genética , Cinética , Camundongos , Camundongos Endogâmicos BALB C , Modelos Químicos , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Estrutura Terciária de Proteína , Eletricidade Estática , Ressonância de Plasmônio de Superfície
17.
J Virol ; 80(6): 2738-46, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16501083

RESUMO

Ebola virus (EBOV) causes a severe hemorrhagic fever for which there are currently no vaccines or effective treatments. While lethal human outbreaks have so far been restricted to sub-Saharan Africa, the potential exploitation of EBOV as a biological weapon cannot be ignored. Two species of EBOV, Sudan ebolavirus (SEBOV) and Zaire ebolavirus (ZEBOV), have been responsible for all of the deadly human outbreaks resulting from this virus. Therefore, it is important to develop a vaccine that can prevent infection by both lethal species. Here, we describe the bivalent cAdVaxE(GPs/z) vaccine, which includes the SEBOV glycoprotein (GP) and ZEBOV GP genes together in a single complex adenovirus-based vaccine (cAdVax) vector. Vaccination of mice with the bivalent cAdVaxE(GPs/z) vaccine led to efficient induction of EBOV-specific antibody and cell-mediated immune responses to both species of EBOV. In addition, the cAdVax technology demonstrated induction of a 100% protective immune response in mice, as all vaccinated C57BL/6 and BALB/c mice survived challenge with a lethal dose of ZEBOV (30,000 times the 50% lethal dose). This study demonstrates the potential efficacy of a bivalent EBOV vaccine based on a cAdVax vaccine vector design.


Assuntos
Adenoviridae/genética , Vacinas contra Ebola/administração & dosagem , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Proteínas do Envelope Viral/imunologia , Adenoviridae/metabolismo , Animais , Anticorpos Antivirais/sangue , Linhagem Celular , Vacinas contra Ebola/genética , Vacinas contra Ebola/imunologia , Ebolavirus/genética , Ebolavirus/imunologia , Ebolavirus/patogenicidade , Células HeLa , Doença pelo Vírus Ebola/virologia , Humanos , Imunidade Celular , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Especificidade da Espécie , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
18.
J Virol ; 79(22): 14189-96, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16254354

RESUMO

Infection with Ebola virus causes a severe disease accompanied by high mortality rates, and there are no licensed vaccines or therapies available for human use. Filovirus vaccine research efforts still need to determine the roles of humoral and cell-mediated immune responses in protection from Ebola virus infection. Previous studies indicated that exposure to Ebola virus proteins expressed from packaged Venezuelan equine encephalitis virus replicons elicited protective immunity in mice and that antibody-mediated protection could only be demonstrated after vaccination against the glycoprotein. In this study, the murine CD8(+) T-cell responses to six Ebola virus proteins were examined. CD8(+) T cells specific for Ebola virus glycoprotein, nucleoprotein, and viral proteins (VP24, VP30, VP35, and VP40) were identified by intracellular cytokine assays using splenocytes from vaccinated mice. The cells were expanded by restimulation with peptides and demonstrated cytolytic activity. Adoptive transfer of the CD8(+) cytotoxic T cells protected filovirus naïve mice from challenge with Ebola virus. These data support a role for CD8(+) cytotoxic T cells as part of a protective mechanism induced by vaccination against six Ebola virus proteins and provide additional evidence that cytotoxic T-cell responses can contribute to protection from filovirus infections.


Assuntos
Ebolavirus/imunologia , Vírus da Encefalite Equina Venezuelana/imunologia , Replicon/imunologia , Linfócitos T Citotóxicos/imunologia , Vacinas Virais , Sequência de Aminoácidos , Animais , Modelos Animais de Doenças , Ebolavirus/genética , Vírus da Encefalite Equina Venezuelana/genética , Encefalomielite Equina Venezuelana/imunologia , Epitopos/química , Ativação Linfocitária , Camundongos , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Replicon/genética , Proteínas Virais/química , Proteínas Virais/imunologia
19.
J Immunol ; 175(2): 1184-91, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16002721

RESUMO

Ebola virus (EBOV)-like particles (eVLP), composed of the EBOV glycoprotein and matrix viral protein (VP)40 with a lipid membrane, are a highly efficacious method of immunization against EBOV infection. The exact requirements for immunity against EBOV infection are poorly defined at this time. The goal of this work was to determine the requirements for EBOV immunity following eVLP vaccination. Vaccination of BALB/c or C57BL/6 mice with eVLPs in conjunction with QS-21 adjuvant resulted in mixed IgG subclass responses, a Th1-like memory cytokine response, and protection from lethal EBOV challenge. Further, this vaccination schedule led to the generation of both CD4(+) and CD8(+) IFN-gamma(+) T cells recognizing specific peptides within glycoprotein and VP40. The transfer of both serum and splenocytes, but not serum or splenocytes alone, from eVLP-vaccinated mice conferred protection against lethal EBOV infection in these studies. B cells were required for eVLP-mediated immunity to EBOV because B cell-deficient mice vaccinated with eVLPs were not protected from lethal EBOV challenge. We also found that CD8(+), but not CD4(+), T cells are absolutely required for eVLP-mediated protection against EBOV infection. Further, eVLP-induced protective mechanisms were perforin-independent, but IFN-gamma-dependent. Taken together, both EBOV-specific humoral and cytotoxic CD8(+) T cell responses are critical to mediate protection against filoviruses following eVLP vaccination.


Assuntos
Anticorpos Antivirais/biossíntese , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Ebolavirus/imunologia , Doença pelo Vírus Ebola/mortalidade , Doença pelo Vírus Ebola/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/fisiologia , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Epitopos de Linfócito T/imunologia , Feminino , Doença pelo Vírus Ebola/imunologia , Interferon gama/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Saponinas/administração & dosagem , Saponinas/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas da Matriz Viral/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Vírion/imunologia
20.
Vaccine ; 21(25-26): 4071-80, 2003 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-12922144

RESUMO

Multiagent DNA vaccines for highly pathogenic organisms offer an attractive approach for preventing naturally occurring or deliberately introduced diseases. Few animal studies have compared the feasibility of combining unrelated gene vaccines. Here, we demonstrate that DNA vaccines to four dissimilar pathogens that are known biowarfare agents, Bacillus anthracis, Ebola (EBOV), Marburg (MARV), and Venezuelan equine encephalitis virus (VEEV), can elicit protective immunity in relevant animal models. In addition, a combination of all four vaccines is shown to be equally as effective as the individual vaccines for eliciting immune responses in a single animal species. These results demonstrate for the first time the potential of combined DNA vaccines for these agents and point to a possible method of rapid development of multiagent vaccines for disparate pathogens such as those that might be encountered in a biological attack.


Assuntos
Vacinas contra Antraz/imunologia , Ebolavirus/imunologia , Vírus da Encefalite Equina Venezuelana/imunologia , Marburgvirus/imunologia , Vacinas Virais/imunologia , Animais , Antraz/imunologia , Antraz/prevenção & controle , Anticorpos Antibacterianos/biossíntese , Anticorpos Antivirais/biossíntese , Biolística , Encefalomielite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/prevenção & controle , Ensaio de Imunoadsorção Enzimática , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Humanos , Macaca fascicularis , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/imunologia , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vacinas Combinadas/imunologia , Vacinas de DNA/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...