Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(15)2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-39126087

RESUMO

Marburg virus (MARV), a filovirus, was first identified in 1967 in Marburg, Germany, and Belgrade, former Yugoslavia. Since then, MARV has caused sporadic outbreaks of human disease with high case fatality rates in parts of Africa, with the largest outbreak occurring in 2004/05 in Angola. From 2021 to 2023, MARV outbreaks occurred in Guinea, Ghana, New Guinea, and Tanzania, emphasizing the expansion of its endemic area into new geographical regions. There are currently no approved vaccines or therapeutics targeting MARV, but several vaccine candidates have shown promise in preclinical studies. We compared three vaccine platforms simultaneously by vaccinating hamsters with either a single dose of an adenovirus-based (ChAdOx-1 MARV) vaccine, an alphavirus replicon-based RNA (LION-MARV) vaccine, or a recombinant vesicular stomatitis virus-based (VSV-MARV) vaccine, all expressing the MARV glycoprotein as the antigen. Lethal challenge with hamster-adapted MARV 4 weeks after vaccination resulted in uniform protection of the VSV-MARV and LION-MARV groups and 83% of the ChAdOx-1 MARV group. Assessment of the antigen-specific humoral response and its functionality revealed vaccine-platform-dependent differences, particularly in the Fc effector functions.


Assuntos
Doença do Vírus de Marburg , Marburgvirus , Vacinas Virais , Animais , Cricetinae , Vacinas Virais/imunologia , Marburgvirus/imunologia , Doença do Vírus de Marburg/prevenção & controle , Doença do Vírus de Marburg/imunologia , Modelos Animais de Doenças , Adenoviridae/genética , Adenoviridae/imunologia , Vesiculovirus/imunologia , Vesiculovirus/genética , Anticorpos Antivirais/imunologia , Vacinação/métodos
2.
J Virol ; 98(7): e0015524, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38832790

RESUMO

Marburg virus infection in humans is associated with case fatality rates that can reach up to 90%, but to date, there are no approved vaccines or monoclonal antibody (mAb) countermeasures. Here, we immunized Rhesus macaques with multivalent combinations of filovirus glycoprotein (GP) antigens belonging to Marburg, Sudan, and Ebola viruses to generate monospecific and cross-reactive antibody responses against them. From the animal that developed the highest titers of Marburg virus GP-specific neutralizing antibodies, we sorted single memory B cells using a heterologous Ravn virus GP probe and cloned and characterized a panel of 34 mAbs belonging to 28 unique lineages. Antibody specificities were assessed by overlapping pepscan and binding competition analyses, revealing that roughly a third of the lineages mapped to the conserved receptor binding region, including potent neutralizing lineages that were confirmed by negative stain electron microscopy to target this region. Additional lineages targeted a protective region on GP2, while others were found to possess cross-filovirus reactivity. Our study advances the understanding of orthomarburgvirus glycoprotein antigenicity and furthers efforts to develop candidate antibody countermeasures against these lethal viruses. IMPORTANCE: Marburg viruses were the first filoviruses characterized to emerge in humans in 1967 and cause severe hemorrhagic fever with average case fatality rates of ~50%. Although mAb countermeasures have been approved for clinical use against the related Ebola viruses, there are currently no approved countermeasures against Marburg viruses. We successfully isolated a panel of orthomarburgvirus GP-specific mAbs from a macaque immunized with a multivalent combination of filovirus antigens. Our analyses revealed that roughly half of the antibodies in the panel mapped to regions on the glycoprotein shown to protect from infection, including the host cell receptor binding domain and a protective region on the membrane-anchoring subunit. Other antibodies in the panel exhibited broad filovirus GP recognition. Our study describes the discovery of a diverse panel of cross-reactive macaque antibodies targeting orthomarburgvirus and other filovirus GPs and provides candidate immunotherapeutics for further study and development.


Assuntos
Anticorpos Monoclonais , Anticorpos Neutralizantes , Anticorpos Antivirais , Reações Cruzadas , Macaca mulatta , Doença do Vírus de Marburg , Marburgvirus , Animais , Marburgvirus/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Monoclonais/imunologia , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/prevenção & controle , Reações Cruzadas/imunologia , Glicoproteínas/imunologia , Proteínas do Envelope Viral/imunologia , Imunização , Humanos , Ebolavirus/imunologia , Antígenos Virais/imunologia
3.
PLoS Pathog ; 20(6): e1012262, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38924060

RESUMO

Viral haemorrhagic fevers (VHF) pose a significant threat to human health. In recent years, VHF outbreaks caused by Ebola, Marburg and Lassa viruses have caused substantial morbidity and mortality in West and Central Africa. In 2022, an Ebola disease outbreak in Uganda caused by Sudan virus resulted in 164 cases with 55 deaths. In 2023, a Marburg disease outbreak was confirmed in Equatorial Guinea and Tanzania resulting in over 49 confirmed or suspected cases; 41 of which were fatal. There are no clearly defined correlates of protection against these VHF, impeding targeted vaccine development. Any vaccine developed should therefore induce strong and preferably long-lasting humoral and cellular immunity against these viruses. Ideally this immunity should also cross-protect against viral variants, which are known to circulate in animal reservoirs and cause human disease. We have utilized two viral vectored vaccine platforms, an adenovirus (ChAdOx1) and Modified Vaccinia Ankara (MVA), to develop a multi-pathogen vaccine regime against three filoviruses (Ebola virus, Sudan virus, Marburg virus) and an arenavirus (Lassa virus). These platform technologies have consistently demonstrated the capability to induce robust cellular and humoral antigen-specific immunity in humans, most recently in the rollout of the licensed ChAdOx1-nCoV19/AZD1222. Here, we show that our multi-pathogen vaccines elicit strong cellular and humoral immunity, induce a diverse range of chemokines and cytokines, and most importantly, confers protection after lethal Ebola virus, Sudan virus and Marburg virus challenges in a small animal model.


Assuntos
Ebolavirus , Doença pelo Vírus Ebola , Febre Lassa , Vírus Lassa , Doença do Vírus de Marburg , Marburgvirus , Animais , Camundongos , Ebolavirus/imunologia , Vírus Lassa/imunologia , Marburgvirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/imunologia , Febre Lassa/imunologia , Febre Lassa/prevenção & controle , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/prevenção & controle , Vacinas Virais/imunologia , Humanos , Vacinação , Feminino , Anticorpos Antivirais/imunologia , Imunogenicidade da Vacina , Vacinas contra Ebola/imunologia
4.
Sci Transl Med ; 14(675): eabq6364, 2022 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-36516269

RESUMO

Marburg virus (MARV) causes a severe hemorrhagic fever disease in primates with mortality rates in humans of up to 90%. MARV has been identified as a category A bioterrorism agent by the Centers for Disease Control and Prevention (CDC) and priority pathogen A by the National Institute of Allergy and Infectious Diseases (NIAID), needing urgent research and development of countermeasures because of the high public health risk it poses. The recent cases of MARV in West Africa underscore the substantial outbreak potential of this virus. The potential for cross-border spread, as had occurred during the 2014-2016 Ebola virus outbreak, illustrates the critical need for MARV vaccines. To support regulatory approval of the chimpanzee adenovirus 3 (ChAd3)-MARV vaccine that has completed phase 1 trials, we showed that the nonreplicating ChAd3 vector, which has a demonstrated safety profile in humans, protected against a uniformly lethal challenge with MARV/Ang. Protective immunity was achieved within 7 days of vaccination and was maintained through 1 year after vaccination. Antigen-specific antibodies were an immune correlate of protection in the acute challenge model, and their concentration was predictive of protection. These results demonstrate that a single-shot ChAd3-MARV vaccine generated a protective immune response that was both rapid and durable with an immune correlate of protection that will support advanced clinical development.


Assuntos
Vacinas contra Ebola , Ebolavirus , Doença pelo Vírus Ebola , Doença do Vírus de Marburg , Marburgvirus , Animais , Humanos , Pan troglodytes , Primatas , Adenoviridae , Doença do Vírus de Marburg/prevenção & controle
6.
Nat Commun ; 12(1): 1891, 2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33767178

RESUMO

Monoclonal antibodies (mAbs) and remdesivir, a small-molecule antiviral, are promising monotherapies for many viruses, including members of the genera Marburgvirus and Ebolavirus (family Filoviridae), and more recently, SARS-CoV-2. One of the major challenges of acute viral infections is the treatment of advanced disease. Thus, extending the window of therapeutic intervention is critical. Here, we explore the benefit of combination therapy with a mAb and remdesivir in a non-human primate model of Marburg virus (MARV) disease. While rhesus monkeys are protected against lethal infection when treatment with either a human mAb (MR186-YTE; 100%), or remdesivir (80%), is initiated 5 days post-inoculation (dpi) with MARV, no animals survive when either treatment is initiated alone beginning 6 dpi. However, by combining MR186-YTE with remdesivir beginning 6 dpi, significant protection (80%) is achieved, thereby extending the therapeutic window. These results suggest value in exploring combination therapy in patients presenting with advanced filovirus disease.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antivirais/uso terapêutico , Doença do Vírus de Marburg/tratamento farmacológico , Marburgvirus/efeitos dos fármacos , Monofosfato de Adenosina/uso terapêutico , Alanina/uso terapêutico , Animais , Modelos Animais de Doenças , Quimioterapia Combinada , Macaca mulatta , Doença do Vírus de Marburg/prevenção & controle , Carga Viral/efeitos dos fármacos
7.
PLoS One ; 13(2): e0192312, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29462200

RESUMO

The search for a universal filovirus vaccine that provides protection against multiple filovirus species has been prompted by sporadic but highly lethal outbreaks of Ebolavirus and Marburgvirus infections. A good prophylactic vaccine should be able to provide protection to all known filovirus species and as an upside potentially protect from newly emerging virus strains. We investigated the immunogenicity and protection elicited by multivalent vaccines expressing glycoproteins (GP) from Ebola virus (EBOV), Sudan virus (SUDV), Taï Forest virus (TAFV) and Marburg virus (MARV). Immune responses against filovirus GP have been associated with protection from disease. The GP antigens were expressed by adenovirus serotypes 26 and 35 (Ad26 and Ad35) and modified Vaccinia virus Ankara (MVA) vectors, all selected for their strong immunogenicity and good safety profile. Using fully lethal NHP intramuscular challenge models, we assessed different vaccination regimens for immunogenicity and protection from filovirus disease. Heterologous multivalent Ad26-Ad35 prime-boost vaccination regimens could give full protection against MARV (range 75%-100% protection) and EBOV (range 50% to 100%) challenge, and partial protection (75%) against SUDV challenge. Heterologous multivalent Ad26-MVA prime-boost immunization gave full protection against EBOV challenge in a small cohort study. The use of such multivalent vaccines did not show overt immune interference in comparison with monovalent vaccines. Multivalent vaccines induced GP-specific antibody responses and cellular IFNγ responses to each GP expressed by the vaccine, and cross-reactivity to TAFV GP was detected in a trivalent vaccine expressing GP from EBOV, SUDV and MARV. In the EBOV challenge studies, higher humoral EBOV GP-specific immune responses (p = 0.0004) were associated with survival from EBOV challenge and less so for cellular immune responses (p = 0.0320). These results demonstrate that it is feasible to generate a multivalent filovirus vaccine that can protect against lethal infection by multiple members of the filovirus family.


Assuntos
Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/imunologia , Vacinas Virais/imunologia , Animais , Feminino , Macaca fascicularis , Masculino
8.
Front Immunol ; 9: 3071, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30723475

RESUMO

Marburg virus (MARV) is the causative agent of hemorrhagic fever outbreaks with high case fatality rates. Closely related to Ebola virus, MARV is a filamentous virus with a negative-sense, single-stranded RNA genome. Although extensive studies on filovirus countermeasures have been conducted, there are no licensed treatments against MARV infections. An experimental vaccine based on the recombinant vesicular stomatitis virus (VSV) expressing the MARV-Musoke glycoprotein demonstrated complete protection when a single dose was administered 28 days and up to 14 months prior to MARV challenge. Here, we analyzed the protective efficacy of an updated vaccine expressing the MARV-Angola glycoprotein (VSV-MARV). A single dose of VSV-MARV given 5 weeks before challenge provided uniform protection with no detectable viremia. The vaccine induced B and T cell proliferation and, importantly, antigen-specific IgG production. Transcriptomic signatures confirm these findings and suggest innate immunity engendered by VSV-MARV may direct the development of protective humoral immunity.


Assuntos
Ativação Linfocitária , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/imunologia , Vacinas Virais/administração & dosagem , Viremia/prevenção & controle , Animais , Linfócitos B/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Macaca fascicularis , Masculino , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/virologia , Linfócitos T/imunologia , Resultado do Tratamento , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vesiculovirus/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Viremia/imunologia , Viremia/virologia
9.
Viral Immunol ; 28(1): 62-70, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25514232

RESUMO

Filoviruses are causative agents of hemorrhagic fever, and to date no effective vaccine or therapeutic has been approved to combat infection. Filovirus glycoprotein (GP) is the critical immunogenic component of filovirus vaccines, eliciting high levels of antibody after successful vaccination. Previous work has shown that protection against both Ebola virus (EBOV) and Marburg virus (MARV) can be achieved by vaccinating with a mixture of virus-like particles (VLPs) expressing either EBOV GP or MARV GP. In this study, the potential for eliciting effective immune responses against EBOV, Sudan virus, and MARV with a single GP construct was tested. Trimeric hybrid GPs were produced that expressed the sequence of Marburg GP2 in conjunction with a hybrid GP1 composed EBOV and Sudan virus GP sequences. VLPs expressing these constructs, along with EBOV VP40, provided comparable protection against MARV challenge, resulting in 75 or 100% protection. Protection from EBOV challenge differed depending upon the hybrid used, however, with one conferring 75% protection and one conferring no protection. By comparing the overall antibody titers and the neutralizing antibody titers specific for each virus, it is shown that higher antibody responses were elicited by the C terminal region of GP1 than by the N terminal region, and this correlated with protection. These data collectively suggest that GP2 and the C terminal region of GP1 are highly immunogenic, and they advance progress toward the development of a pan-filovirus vaccine.


Assuntos
Proteção Cruzada , Ebolavirus/imunologia , Marburgvirus/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Antígenos Virais/genética , Antígenos Virais/imunologia , Ebolavirus/genética , Feminino , Cobaias , Doença pelo Vírus Ebola/prevenção & controle , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Análise de Sobrevida , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/genética , Proteínas do Envelope Viral/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Virossomos/genética , Virossomos/imunologia
11.
Nature ; 508(7496): 402-5, 2014 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-24590073

RESUMO

Filoviruses are emerging pathogens and causative agents of viral haemorrhagic fever. Case fatality rates of filovirus disease outbreaks are among the highest reported for any human pathogen, exceeding 90% (ref. 1). Licensed therapeutic or vaccine products are not available to treat filovirus diseases. Candidate therapeutics previously shown to be efficacious in non-human primate disease models are based on virus-specific designs and have limited broad-spectrum antiviral potential. Here we show that BCX4430, a novel synthetic adenosine analogue, inhibits infection of distinct filoviruses in human cells. Biochemical, reporter-based and primer-extension assays indicate that BCX4430 inhibits viral RNA polymerase function, acting as a non-obligate RNA chain terminator. Post-exposure intramuscular administration of BCX4430 protects against Ebola virus and Marburg virus disease in rodent models. Most importantly, BCX4430 completely protects cynomolgus macaques from Marburg virus infection when administered as late as 48 hours after infection. In addition, BCX4430 exhibits broad-spectrum antiviral activity against numerous viruses, including bunyaviruses, arenaviruses, paramyxoviruses, coronaviruses and flaviviruses. This is the first report, to our knowledge, of non-human primate protection from filovirus disease by a synthetic drug-like small molecule. We provide additional pharmacological characterizations supporting the potential development of BCX4430 as a countermeasure against human filovirus diseases and other viral diseases representing major public health threats.


Assuntos
Adenosina/análogos & derivados , Antivirais/farmacologia , Infecções por Filoviridae/prevenção & controle , Infecções por Filoviridae/virologia , Filoviridae/efeitos dos fármacos , Nucleosídeos de Purina/farmacologia , Adenina/análogos & derivados , Administração Oral , Animais , Antivirais/administração & dosagem , Antivirais/química , Antivirais/farmacocinética , RNA Polimerases Dirigidas por DNA/antagonistas & inibidores , RNA Polimerases Dirigidas por DNA/metabolismo , Modelos Animais de Doenças , Ebolavirus/efeitos dos fármacos , Filoviridae/enzimologia , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/virologia , Humanos , Injeções Intramusculares , Macaca fascicularis/virologia , Doença do Vírus de Marburg/prevenção & controle , Doença do Vírus de Marburg/virologia , Marburgvirus/efeitos dos fármacos , Nucleosídeos de Purina/administração & dosagem , Nucleosídeos de Purina/química , Nucleosídeos de Purina/farmacocinética , Pirrolidinas , RNA/biossíntese , Fatores de Tempo
12.
Virus Res ; 162(1-2): 148-61, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21925552

RESUMO

Developing vaccines for highly pathogenic viruses such as those causing Lassa, Ebola, and Marburg hemorrhagic fevers is a daunting task due to both scientific and logistical constraints. Scientific hurdles to overcome include poorly defined relationships between pathogenicity and protective immune responses, genetic diversity of viruses, and safety in a target population that includes a large number of individuals with compromised immune systems. Logistical obstacles include the requirement for biosafety level-4 containment to study the authentic viruses, the poor public health infrastructure of the endemic disease areas, and the cost of developing these vaccines for use in non-lucrative markets. Recombinant DNA-based vaccine approaches offer promise of overcoming some of these issues. In this review, we consider the status of various recombinant DNA candidate vaccines against Lassa virus and filoviruses which have been tested in animals.


Assuntos
Ebolavirus/genética , Doença pelo Vírus Ebola/prevenção & controle , Febre Lassa/prevenção & controle , Vírus Lassa/genética , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/genética , Vacinação , Vacinas de DNA/genética , Adenoviridae/imunologia , Animais , Ebolavirus/imunologia , Variação Genética , Vetores Genéticos/imunologia , Doença pelo Vírus Ebola/imunologia , Humanos , Febre Lassa/imunologia , Vírus Lassa/imunologia , Doença do Vírus de Marburg/imunologia , Marburgvirus/imunologia , Plasmídeos/imunologia , Saúde Pública , Vacinas de DNA/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas de Partículas Semelhantes a Vírus/genética , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vesiculovirus/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia , Replicação Viral/genética
13.
Expert Rev Vaccines ; 10(1): 63-77, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21162622

RESUMO

Marburg and Ebola viruses cause severe hemorrhagic fever in humans and nonhuman primates. Currently, there are no effective treatments and no licensed vaccines; although a number of vaccine platforms have proven successful in animal models. The ideal filovirus vaccine candidate should be able to provide rapid protection following a single immunization, have the potential to work postexposure and be cross-reactive or multivalent against all Marburg virus strains and all relevant Ebola virus species and strains. Currently, there are multiple platforms that have provided prophylactic protection in nonhuman primates, including DNA, recombinant adenovirus serotype 5, recombinant human parainfluenza virus 3 and virus-like particles. In addition, a single platform, recombinant vesicular stomatitis virus, has demonstrated both prophylactic and postexposure protection in nonhuman primates. These results demonstrate that achieving a vaccine that is protective against filoviruses is possible; the challenge now is to prove its safety and efficacy in order to obtain a vaccine that is ready for human use.


Assuntos
Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/imunologia , Vacinas Virais/imunologia , Adenoviridae/genética , Animais , Modelos Animais de Doenças , Portadores de Fármacos , Ebolavirus/genética , Vetores Genéticos , Humanos , Marburgvirus/genética , Vírus da Parainfluenza 3 Humana/genética , Primatas , Vacinas de DNA/efeitos adversos , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vesiculovirus/genética , Vacinas Virais/efeitos adversos , Vacinas Virais/genética
14.
J Virol ; 84(19): 10386-94, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20660192

RESUMO

The immunogenicity and durability of genetic vaccines are influenced by the composition of gene inserts and choice of delivery vector. DNA vectors are a promising vaccine approach showing efficacy when combined in prime-boost regimens with recombinant protein or viral vectors, but they have shown limited comparative efficacy as a stand-alone platform in primates, due possibly to suboptimal gene expression or cell targeting. Here, regimens using DNA plasmids modified for optimal antigen expression and recombinant adenovirus (rAd) vectors, all encoding the glycoprotein (GP) gene from Angola Marburg virus (MARV), were compared for their ability to provide immune protection against lethal MARV Angola infection. Heterologous DNA-GP/rAd5-GP prime-boost and single-modality rAd5-GP, as well as the DNA-GP-only vaccine, prevented death in all vaccinated subjects after challenge with a lethal dose of MARV Angola. The DNA/DNA vaccine induced humoral responses comparable to those induced by a single inoculation with rAd5-GP, as well as CD4(+) and CD8(+) cellular immune responses, with skewing toward CD4(+) T-cell activity against MARV GP. Vaccine regimens containing rAd-GP, alone or as a boost, exhibited cellular responses with CD8(+) T-cell dominance. Across vaccine groups, CD8(+) T-cell subset dominance comprising cells exhibiting a tumor necrosis factor alpha (TNF-alpha) and gamma interferon (IFN-gamma) double-positive functional phenotype was associated with an absence or low frequency of clinical symptoms, suggesting that both the magnitude and functional phenotype of CD8(+) T cells may determine vaccine efficacy against infection by MARV Angola.


Assuntos
Vetores Genéticos , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/genética , Marburgvirus/imunologia , Vacinas Virais/farmacologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/metabolismo , Genes Virais , Imunidade Celular , Imunização Secundária , Macaca fascicularis , Modelos Animais , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Vacinas de DNA/farmacologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia
15.
Rev Med Virol ; 20(6): 344-57, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20658513

RESUMO

For more than 30 years the filoviruses, Marburg virus and Ebola virus, have been associated with periodic outbreaks of hemorrhagic fever that produce severe and often fatal disease. The filoviruses are endemic primarily in resource-poor regions in Central Africa and are also potential agents of bioterrorism. Although no vaccines or antiviral drugs for Marburg or Ebola are currently available, remarkable progress has been made over the last decade in developing candidate preventive vaccines against filoviruses in nonhuman primate models. Due to the generally remote locations of filovirus outbreaks, a single-injection vaccine is desirable. Among the prospective vaccines that have shown efficacy in nonhuman primate models of filoviral hemorrhagic fever, two candidates, one based on a replication-defective adenovirus serotype 5 and the other on a recombinant VSV (rVSV), were shown to provide complete protection to nonhuman primates when administered as a single injection. The rVSV-based vaccine has also shown utility when administered for postexposure prophylaxis against filovirus infections. A VSV-based Ebola vaccine was recently used to manage a potential laboratory exposure.


Assuntos
Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/imunologia , Vacinas Virais/imunologia , Adenoviridae/genética , Animais , Modelos Animais de Doenças , Vetores Genéticos , Doença pelo Vírus Ebola/epidemiologia , Humanos , Doença do Vírus de Marburg/epidemiologia , Primatas , Vacinação/métodos , Vacinas Sintéticas/imunologia , Vesiculovirus/genética
16.
Virol J ; 6: 132, 2009 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-19712478

RESUMO

BACKGROUND: Marburg virus (MARV) causes acute hemorrhagic fever that is often lethal, and no licensed vaccines are available for preventing this deadly viral infection. The immune mechanisms for protection against MARV are poorly understood, but previous studies suggest that both antibodies and T cells are required. In our study, we infected BALB/c mice with plaque-purified, nonlethal MARV and used overlapping peptides to map H2d-restricted CD8+ T-cell epitopes. METHODS: Splenocytes from mice infected with nonlethal MARV were harvested and stimulated with multiple overlapping 15-mer peptide pools, and reactive CD8+ T cells were evaluated for antigen specificity by measuring upregulation of CD44 and interferon-gamma expression. After confirming positive reactivity to specific 15-mer peptides, we used extrapolated 9-mer epitopes to evaluate the induction of cytotoxic T-cell responses and protection from lethal MARV challenge in BALB/c mice. RESULTS: We discovered a CD8+ T-cell epitope within both the MARV glycoprotein (GP) and nucleoprotein (NP) that triggered cytotoxic T-cell responses. These responses were also protective when epitope-specific splenocytes were transferred into naïve animals. CONCLUSION: Epitope mapping of MARV GP, NP, and VP40 provides the first evidence that specific MARV-epitope induction of cellular immune responses is sufficient to combat infection. Establishment of CD8+ T-cell epitopes that are reactive to MARV proteins provides an important research tool for dissecting the significance of cellular immune responses in BALB/c mice infected with MARV.


Assuntos
Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Doença do Vírus de Marburg/imunologia , Marburgvirus/imunologia , Sequência de Aminoácidos , Animais , Linfócitos T CD8-Positivos/química , Linfócitos T CD8-Positivos/imunologia , Mapeamento de Epitopos , Epitopos de Linfócito T/química , Humanos , Doença do Vírus de Marburg/prevenção & controle , Doença do Vírus de Marburg/virologia , Marburgvirus/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Dados de Sequência Molecular , Nucleoproteínas/química , Nucleoproteínas/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia
17.
J Immunotoxicol ; 5(3): 315-35, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18830892

RESUMO

The Vaccine Research Center has developed vaccine candidates for different diseases/infectious agents (including HIV-1, Ebola, and Marburg viruses) built on an adenovirus vector platform, based on adenovirus type 5 or 35. To support clinical development of each vaccine candidate, pre-clinical studies were performed in rabbits to determine where in the body they biodistribute and how rapidly they clear, and to screen for potential toxicities (intrinsic and immunotoxicities). The vaccines biodistribute only to spleen, liver (Ad5 only), and/or iliac lymph node (Ad35 only) and otherwise remain in the site of injection muscle and overlying subcutis. Though approximately 10(11) viral particles were inoculated, already by Day 9, all but 10(3) to 10(5) genome copies per mu g of DNA had cleared from the injection site muscle. By three months, the adenovector was cleared with, at most, a few animals retaining a small number of copies in the injection site, spleen (Ad5), or iliac lymph node (Ad35). This pattern of limited biodistribution and extensive clearance is consistent regardless of differences in adenovector type (Ad5 or 35), manufacturer's construct and production methods, or gene-insert. Repeated dose toxicology studies identified treatment-related toxicities confined primarily to the sites of injection, in certain clinical pathology parameters, and in body temperatures (Ad5 vectors) and food consumption immediately post-inoculation. Systemic reactogenicity and reactogenicity at the sites of injection demonstrated reversibility. These data demonstrate the safety and suitability for investigational human use of Ad5 or Ad35 adenovector-based vaccine candidates at doses of up to 2 x 10(11) given intramuscularly to prevent various infectious diseases.


Assuntos
Vacinas contra a AIDS/farmacocinética , Vacinas contra Ebola/farmacocinética , Ebolavirus/imunologia , HIV-1/imunologia , Marburgvirus/imunologia , Vacinas Virais/farmacocinética , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/imunologia , Vacinas contra a AIDS/toxicidade , Adenoviridae/classificação , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Anticorpos Antivirais/sangue , DNA Viral/análise , Avaliação Pré-Clínica de Medicamentos , Vacinas contra Ebola/administração & dosagem , Vacinas contra Ebola/imunologia , Vacinas contra Ebola/toxicidade , Feminino , Vetores Genéticos/classificação , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Vetores Genéticos/farmacocinética , Infecções por HIV/prevenção & controle , Doença pelo Vírus Ebola/prevenção & controle , Humanos , Injeções Intramusculares , Masculino , Doença do Vírus de Marburg/prevenção & controle , Reação em Cadeia da Polimerase , Coelhos , Sorotipagem , Fatores de Tempo , Distribuição Tecidual , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Vacinas de DNA/farmacocinética , Vacinas de DNA/toxicidade , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Vacinas Virais/toxicidade
18.
Clin Vaccine Immunol ; 15(3): 460-7, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18216185

RESUMO

Filoviruses (Ebola and Marburg viruses) are among the deadliest viruses known to mankind, with mortality rates nearing 90%. These pathogens are highly infectious through contact with infected body fluids and can be easily aerosolized. Additionally, there are currently no licensed vaccines available to prevent filovirus outbreaks. Their high mortality rates and infectious capabilities when aerosolized and the lack of licensed vaccines available to prevent such infectious make Ebola and Marburg viruses serious bioterrorism threats, placing them both on the category A list of bioterrorism agents. Here we describe a panfilovirus vaccine based on a complex adenovirus (CAdVax) technology that expresses multiple antigens from five different filoviruses de novo. Vaccination of nonhuman primates demonstrated 100% protection against infection by two species of Ebola virus and three Marburg virus subtypes, each administered at 1,000 times the lethal dose. This study indicates the feasibility of vaccination against all current filovirus threats in the event of natural hemorrhagic fever outbreak or biological attack.


Assuntos
Adenoviridae/genética , Vacinas contra Ebola , Filoviridae , Vetores Genéticos , Doença pelo Vírus Ebola/prevenção & controle , Doença do Vírus de Marburg/prevenção & controle , Vacinas Virais , Adenoviridae/imunologia , Animais , Antígenos Virais/genética , Antígenos Virais/metabolismo , Bioterrorismo/prevenção & controle , Vacinas contra Ebola/administração & dosagem , Vacinas contra Ebola/genética , Vacinas contra Ebola/imunologia , Ebolavirus/imunologia , Ebolavirus/patogenicidade , Filoviridae/classificação , Filoviridae/genética , Filoviridae/imunologia , Doença pelo Vírus Ebola/imunologia , Humanos , Macaca fascicularis , Doença do Vírus de Marburg/imunologia , Marburgvirus/classificação , Marburgvirus/imunologia , Marburgvirus/patogenicidade , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia
19.
J Infect Dis ; 196 Suppl 2: S168-75, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17940946

RESUMO

BACKGROUND: Occupational transmission to health workers (HWs) has been a typical feature of Marburg hemorrhagic fever (MHF) outbreaks. The goal of this study was to identify cases of occupational MHF in HWs from Durba and Watsa, Democratic Republic of the Congo; to assess levels of exposure and protection; and to explore reasons for inconsistent use of protective gear. METHODS: A serosurvey of 48 HWs who cared for patients with MHF was performed. In addition, HWs were given a questionnaire on types of exposure, use of protective gear, and symptoms after contact. Informal and in-depth interviews with HWs were also performed. RESULTS: We found 1 HW who was seropositive for MHF, in addition to 5 cases of occupational MHF known beforehand; 4 infections had occurred after the introduction of infection control. HWs protected themselves better during invasive procedures (injections, venipuncture, and surgery) than during noninvasive procedures, but the overall level of protection in the hospital remained insufficient, particularly outside of isolation wards. The reasons for inconsistent use of protective gear included insufficient availability of the gear, adherence to traditional explanatory models of the origin of disease, and peer bonding with sick colleagues. CONCLUSIONS: Infection control must not focus too exclusively on the establishment of isolation wards but should aim at improving overall hospital hygiene. Training of HWs should allow them to voice and discuss their doubts and prepare them for the peculiarities of caring for ill colleagues.


Assuntos
Pessoal de Saúde , Doença do Vírus de Marburg/epidemiologia , Roupa de Proteção , Animais , República Democrática do Congo/epidemiologia , Surtos de Doenças , Luvas Protetoras , Pessoal de Saúde/estatística & dados numéricos , Humanos , Entrevistas como Assunto , Doença do Vírus de Marburg/prevenção & controle , Doença do Vírus de Marburg/transmissão , Doenças Profissionais/prevenção & controle , Doenças Profissionais/virologia
20.
Virology ; 353(2): 324-32, 2006 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-16820184

RESUMO

The Marburg virus (MARV), an African filovirus closely related to the Ebola virus, causes a deadly hemorrhagic fever in humans, with up to 90% mortality. Currently, treatment of disease is only supportive, and no vaccines are available to prevent spread of MARV infections. In order to address this need, we have developed and characterized a novel recombinant vaccine that utilizes a single complex adenovirus-vectored vaccine (cAdVax) to overexpress a MARV glycoprotein (GP) fusion protein derived from the Musoke and Ci67 strains of MARV. Vaccination with the cAdVaxM(fus) vaccine led to efficient production of MARV-specific antibodies in both mice and guinea pigs. Significantly, guinea pigs vaccinated with at least 5 x 10(7) pfu of cAdVaxM(fus) vaccine were 100% protected against lethal challenges by the Musoke, Ci67 and Ravn strains of MARV, making it a vaccine with trivalent protective efficacy. Therefore, the cAdVaxM(fus) vaccine serves as a promising vaccine candidate to prevent and contain multi-strain infections by MARV.


Assuntos
Adenoviridae/metabolismo , Antígenos Virais/biossíntese , Vacinas contra Ebola/administração & dosagem , Vetores Genéticos/metabolismo , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/imunologia , Vacinação , Proteínas do Envelope Viral/biossíntese , Proteínas Virais de Fusão/biossíntese , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/genética , Linhagem Celular , Chlorocebus aethiops , Relação Dose-Resposta Imunológica , Vacinas contra Ebola/genética , Terapia Genética/métodos , Cobaias , Humanos , Injeções Intraperitoneais , Injeções Subcutâneas , Doença do Vírus de Marburg/sangue , Doença do Vírus de Marburg/imunologia , Camundongos , Dados de Sequência Molecular , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Alinhamento de Sequência , Vacinas Sintéticas/administração & dosagem , Proteínas do Envelope Viral/genética , Proteínas Virais de Fusão/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA