Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 17(10): e1009966, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34634087

RESUMO

Nigeria continues to experience ever increasing annual outbreaks of Lassa fever (LF). The World Health Organization has recently declared Lassa virus (LASV) as a priority pathogen for accelerated research leading to a renewed international effort to develop relevant animal models of disease and effective countermeasures to reduce LF morbidity and mortality in endemic West African countries. A limiting factor in evaluating medical countermeasures against LF is a lack of well characterized animal models outside of those based on infection with LASV strain Josiah originating form Sierra Leone, circa 1976. Here we genetically characterize five recent LASV isolates collected from the 2018 outbreak in Nigeria. Three isolates were further evaluated in vivo and despite being closely related and from the same spatial / geographic region of Nigeria, only one of the three isolates proved lethal in strain 13 guinea pigs and non-human primates (NHP). Additionally, this isolate exhibited atypical pathogenesis characteristics in the NHP model, most notably respiratory failure, not commonly described in hemorrhagic cases of LF. These results suggest that there is considerable phenotypic heterogeneity in LASV infections in Nigeria, which leads to a multitude of pathogenesis characteristics that could account for differences between subclinical and lethal LF infections. Most importantly, the development of disease models using currently circulating LASV strains in West Africa are critical for the evaluation of potential vaccines and medical countermeasures.


Assuntos
Modelos Animais de Doenças , Febre Lassa/genética , Vírus Lassa/genética , Animais , Surtos de Doenças , Feminino , Cobaias , Humanos , Macaca fascicularis , Masculino , Nigéria , Filogenia
2.
Emerg Infect Dis ; 26(12): 3020-3024, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33219792

RESUMO

Hantavirus cardiopulmonary syndrome (HCPS) is a severe respiratory disease caused by Sin Nombre virus in North America (SNV). As of January 1, 2020, SNV has caused 143 laboratory-confirmed cases of HCPS in Canada. We review critical aspects of SNV virus epidemiology and the ecology, biology, and genetics of HCPS in Canada.


Assuntos
Infecções por Hantavirus , Síndrome Pulmonar por Hantavirus , Orthohantavírus , Vírus Sin Nombre , Canadá/epidemiologia , Orthohantavírus/genética , Infecções por Hantavirus/epidemiologia , Síndrome Pulmonar por Hantavirus/diagnóstico , Síndrome Pulmonar por Hantavirus/epidemiologia , Humanos , América do Norte
3.
CMAJ ; 195(22): E800-E801, 2023 06 05.
Artigo em Francês | MEDLINE | ID: mdl-37277129
5.
CMAJ ; 195(8): E305, 2023 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-36849175
6.
J Infect Dis ; 214(suppl 3): S218-S221, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27571899

RESUMO

Personnel deployed to remote areas during infectious disease outbreaks have limited access to mechanical and chemical inactivation resources. The inactivation of infectious agents present in diagnostic samples is critical to ensure the safety of personnel and the containment of the disease. We evaluated the efficacy of thermal inactivation (exposure to 56°C for 1 hour) and chemical inactivation with 0.5% Tween-20 against a high titer of Ebola virus (species Zaire ebolavirus) variant Makona in spiked human serum samples. No surviving virus was revealed by a 50% tissue culture infective dose assay after the combined treatment under laboratory conditions. In-field use of this inactivation protocol during the 2013-2016 West Africa Ebola outbreaks demonstrated readily detectable levels of immunoglobulin G and/or immunoglobulin M in human plasma samples after treatment.


Assuntos
Anticorpos Antivirais/sangue , Antígenos Virais/imunologia , Surtos de Doenças , Ebolavirus/imunologia , Doença pelo Vírus Ebola/diagnóstico , Inativação de Vírus , África Ocidental/epidemiologia , Animais , Chlorocebus aethiops , República Democrática do Congo/epidemiologia , Ebolavirus/isolamento & purificação , Ensaio de Imunoadsorção Enzimática , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/virologia , Temperatura Alta , Humanos , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Células Vero
7.
J Infect Dis ; 214(suppl 3): S169-S176, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27333914

RESUMO

West Africa experienced the first epidemic of Ebola virus infection, with by far the greatest number of cases in Guinea, Sierra Leone, and Liberia. The unprecedented epidemic triggered an unparalleled response, including the deployment of multiple Ebola treatment units and mobile/field diagnostic laboratories. The National Institute of Allergy and Infectious Diseases and the Centers for Disease Control and Prevention deployed a joint laboratory to Monrovia, Liberia, in August 2014 to support the newly founded Ebola treatment unit at the Eternal Love Winning Africa (ELWA) campus. The laboratory operated initially out of a tent structure but quickly moved into a fixed-wall building owing to severe weather conditions, the need for increased security, and the high sample volume. Until May 2015, when the laboratory closed, the site handled close to 6000 clinical specimens for Ebola virus diagnosis and supported the medical staff in case patient management. Laboratory operation and safety, as well as Ebola virus diagnostic assays, are described and discussed; in addition, lessons learned for future deployments are reviewed.


Assuntos
Serviços de Laboratório Clínico/organização & administração , Ebolavirus/isolamento & purificação , Epidemias/prevenção & controle , Doença pelo Vírus Ebola/epidemiologia , África Ocidental/epidemiologia , Centers for Disease Control and Prevention, U.S. , Feminino , Guiné/epidemiologia , Doença pelo Vírus Ebola/diagnóstico , Doença pelo Vírus Ebola/transmissão , Doença pelo Vírus Ebola/virologia , Humanos , Cooperação Internacional , Libéria/epidemiologia , Masculino , National Institute of Allergy and Infectious Diseases (U.S.) , Segurança , Serra Leoa/epidemiologia , Estados Unidos
8.
Clin Infect Dis ; 63(8): 1026-33, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27531847

RESUMO

BACKGROUND: The ongoing Ebola outbreak in West Africa has resulted in 28 646 suspected, probable, and confirmed Ebola virus infections. Nevertheless, malaria remains a large public health burden in the region affected by the outbreak. A joint Centers for Disease Control and Prevention/National Institutes of Health diagnostic laboratory was established in Monrovia, Liberia, in August 2014, to provide laboratory diagnostics for Ebola virus. METHODS: All blood samples from suspected Ebola virus-infected patients admitted to the Médecins Sans Frontières ELWA3 Ebola treatment unit in Monrovia were tested by quantitative real-time polymerase chain reaction for the presence of Ebola virus and Plasmodium species RNA. Clinical outcome in laboratory-confirmed Ebola virus-infected patients was analyzed as a function of age, sex, Ebola viremia, and Plasmodium species parasitemia. RESULTS: The case fatality rate of 1182 patients with laboratory-confirmed Ebola virus infections was 52%. The probability of surviving decreased with increasing age and decreased with increasing Ebola viral load. Ebola virus-infected patients were 20% more likely to survive when Plasmodium species parasitemia was detected, even after controlling for Ebola viral load and age; those with the highest levels of parasitemia had a survival rate of 83%. This effect was independent of treatment with antimalarials, as this was provided to all patients. Moreover, treatment with antimalarials did not affect survival in the Ebola virus mouse model. CONCLUSIONS: Plasmodium species parasitemia is associated with an increase in the probability of surviving Ebola virus infection. More research is needed to understand the molecular mechanism underlying this remarkable phenomenon and translate it into treatment options for Ebola virus infection.


Assuntos
Coinfecção , Ebolavirus , Doença pelo Vírus Ebola/complicações , Doença pelo Vírus Ebola/mortalidade , Malária/complicações , Malária/parasitologia , Parasitemia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Criança , Pré-Escolar , Modelos Animais de Doenças , Ebolavirus/genética , Feminino , Doença pelo Vírus Ebola/diagnóstico , Doença pelo Vírus Ebola/epidemiologia , Humanos , Lactente , Recém-Nascido , Malária/diagnóstico , Malária/epidemiologia , Masculino , Camundongos , Pessoa de Meia-Idade , Carga Parasitária , Plasmodium/genética , Taxa de Sobrevida , Carga Viral , Adulto Jovem
9.
Emerg Infect Dis ; 22(2): 323-6, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26814608

RESUMO

Malaria is a major public health concern in the countries affected by the Ebola virus disease epidemic in West Africa. We determined the feasibility of using molecular malaria diagnostics during an Ebola virus disease outbreak and report the incidence of Plasmodium spp. parasitemia in persons with suspected Ebola virus infection.


Assuntos
Coinfecção , Surtos de Doenças , Ebolavirus , Doença pelo Vírus Ebola/epidemiologia , Malária/diagnóstico , Malária/parasitologia , Humanos , Malária Falciparum/diagnóstico , Malária Falciparum/parasitologia , Carga Parasitária , Plasmodium falciparum/classificação , Plasmodium falciparum/genética , Prevalência
10.
J Infect Dis ; 212(11): 1752-8, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26002981

RESUMO

This paper describes patient characteristics, including Ebola viral load, associated with mortality in a Médecins Sans Frontières Ebola case management centre (CMC).Out of 780 admissions between June and October 2014, 525 (67%) were positive for Ebola with a known outcome. The crude mortality rate was 51% (270/525). Ebola viral load (whole-blood sample) data were available on 76% (397/525) of patients. Univariate analysis indicated viral load at admission, age, symptom duration prior to admission, and distance traveled to the CMC were associated with mortality (P < .05). The multivariable model predicted mortality in those with a viral load at admission greater than 10 million copies per milliliter (P < .05, odds ratio >10), aged ≥ 50 years (P = .08, odds ratio = 2) and symptom duration prior to admission less than 5 days (P = .14). The presence of confusion, diarrhea, and conjunctivitis were significantly higher (P < .05) in Ebola patients who died.These findings highlight the importance viral load at admission has on mortality outcomes and could be used to cohort cases with viral loads greater than 10 million copies into dedicated wards with more intensive medical support to further reduce mortality.


Assuntos
Surtos de Doenças/estatística & dados numéricos , Ebolavirus/genética , Doença pelo Vírus Ebola/mortalidade , Doença pelo Vírus Ebola/virologia , Hospitalização/estatística & dados numéricos , Carga Viral/estatística & dados numéricos , Adolescente , Adulto , Feminino , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/terapia , Humanos , Masculino , Reação em Cadeia da Polimerase , Estudos Retrospectivos , Serra Leoa/epidemiologia , Adulto Jovem
11.
Nat Med ; 11(7): 786-90, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15937495

RESUMO

Vaccines and therapies are urgently needed to address public health needs stemming from emerging pathogens and biological threat agents such as the filoviruses Ebola virus (EBOV) and Marburg virus (MARV). Here, we developed replication-competent vaccines against EBOV and MARV based on attenuated recombinant vesicular stomatitis virus vectors expressing either the EBOV glycoprotein or MARV glycoprotein. A single intramuscular injection of the EBOV or MARV vaccine elicited completely protective immune responses in nonhuman primates against lethal EBOV or MARV challenges. Notably, vaccine vector shedding was not detectable in the monkeys and none of the animals developed fever or other symptoms of illness associated with vaccination. The EBOV vaccine induced humoral and apparent cellular immune responses in all vaccinated monkeys, whereas the MARV vaccine induced a stronger humoral than cellular immune response. No evidence of EBOV or MARV replication was detected in any of the protected animals after challenge. Our data suggest that these vaccine candidates are safe and highly efficacious in a relevant animal model.


Assuntos
Ebolavirus/imunologia , Marburgvirus/imunologia , Vacinas Atenuadas/imunologia , Vacinas Combinadas/imunologia , Vacinas Virais/imunologia , Animais , Formação de Anticorpos , Reações Cruzadas , Vacinas contra Ebola/imunologia , Vacinas contra Ebola/farmacologia , Primatas , Vacinas Atenuadas/genética , Vacinas Atenuadas/farmacologia , Vacinas Combinadas/genética , Vacinas Combinadas/farmacologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/farmacologia , Vírus da Estomatite Vesicular Indiana/genética , Vacinas Virais/genética , Vacinas Virais/farmacologia , Viremia/imunologia , Viremia/virologia , Replicação Viral
12.
J Infect Dis ; 204 Suppl 3: S1082-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21987745

RESUMO

The recombinant vesicular stomatitis virus (rVSV) vector-based monovalent vaccine platform expressing a filovirus glycoprotein has been demonstrated to provide protection from lethal challenge with Ebola (EBOV) and Marburg (MARV) viruses both prophylactically and after exposure. This platform provides protection between heterologous strains within a species; however, protection from lethal challenge between species has been largely unsuccessful. To determine whether the rVSV-EBOV vaccines have the potential to provide protection against a newly emerging, phylogenetically related species, cynomolgus macaques were vaccinated with an rVSV vaccine expressing either the glycoprotein of Zaire ebolavirus (ZEBOV) or Côte d'Ivoire ebolavirus (CIEBOV) and then challenged with Bundibugyo ebolavirus (BEBOV), which was recently proposed as a new EBOV species following an outbreak in Uganda in 2007. A single vaccination with the ZEBOV-specific vaccine provided cross-protection (75% survival) against subsequent BEBOV challenge, whereas vaccination with the CIEBOV-specific vaccine resulted in an outcome similar to mock-immunized animals (33% and 25% survival, respectively). This demonstrates that monovalent rVSV-based vaccines may be useful against a newly emerging species; however, heterologous protection across species remains challenging and may depend on enhancing the immune responses either through booster immunizations or through the inclusion of multiple immunogens.


Assuntos
Vacinas contra Ebola/administração & dosagem , Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Vesiculovirus , Animais , Anticorpos Antivirais/sangue , Ebolavirus/classificação , Ebolavirus/genética , Feminino , Macaca fascicularis , Masculino , Projetos Piloto , Especificidade da Espécie , Vacinas Sintéticas
13.
Proc Natl Acad Sci U S A ; 105(46): 17982-7, 2008 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-18981410

RESUMO

Human infections with Ebola virus (EBOV) result in a deadly viral disease known as Ebola hemorrhagic fever. Up to 90% of infected patients die, and there is no available treatment or vaccine. The sporadic human outbreaks are believed to result when EBOV "jumps" from an infected animal to a person and is subsequently transmitted between persons by direct contact with infected blood or body fluids. This study was undertaken to investigate the mechanism by which EBOV can persistently infect and then escape from model cell and animal reservoir systems. We report a model system in which infection of mouse and bat cell lines with EBOV leads to persistence, which can be broken with low levels of lipopolysaccharide or phorbol-12-myristate-13-acetate (PMA). This reactivation depends on the Ras/MAPK pathway through inhibition of RNA-dependent protein kinase and eukaryotic initiation factor 2alpha phosphorylation and occurs at the level of protein synthesis. EBOV also can be evoked from mice 7 days after infection by PMA treatment, indicating that a similar mechanism occurs in vivo. Our findings suggest that EBOV may persist in nature through subclinical infection of a reservoir species, such as bats, and that appropriate physiological stimulation may result in increased replication and transmission to new hosts. Identification of a presumptive mechanism responsible for EBOV emergence from its reservoir underscores the "hit-and-run" nature of the initiation of human and/or nonhuman primate EBOV outbreaks and may provide insight into possible countermeasures to interfere with transmission.


Assuntos
Ebolavirus/metabolismo , Doença pelo Vírus Ebola/enzimologia , Doença pelo Vírus Ebola/virologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas ras/metabolismo , Animais , Linhagem Celular Transformada , Quirópteros , Relação Dose-Resposta a Droga , Ebolavirus/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Fator de Iniciação 2 em Eucariotos/metabolismo , Feminino , Proteínas de Fluorescência Verde/metabolismo , Lipopolissacarídeos/farmacologia , Fígado/virologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Fosforilação/efeitos dos fármacos , Baço/virologia , Acetato de Tetradecanoilforbol/farmacologia
14.
Microorganisms ; 9(3)2021 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-33652895

RESUMO

BACKGROUND: The 2014-2016 Ebola outbreak in West Africa recapitulated that nosocomial spread of Ebola virus could occur and that health care workers were at particular risk including notable cases in Europe and North America. These instances highlighted the need for centers to better prepare for potential Ebola virus cases; including understanding how the virus spreads and which interventions pose the greatest risk. METHODS: We created a fully equipped intensive care unit (ICU), within a Biosafety Level 4 (BSL4) laboratory, and infected multiple sedated non-human primates (NHPs) with Ebola virus. While providing bedside care, we sampled blood, urine, and gastric residuals; as well as buccal, ocular, nasal, rectal, and skin swabs, to assess the risks associated with routine care. We also assessed the physical environment at end-point. RESULTS: Although viral RNA was detectable in blood as early as three days post-infection, it was not detectable in the urine, gastric fluid, or swabs until late-stage disease. While droplet spread and fomite contamination were present on a few of the surfaces that were routinely touched while providing care in the ICU for the infected animal, these may have been abrogated through good routine hygiene practices. CONCLUSIONS: Overall this study has helped further our understanding of which procedures may pose the highest risk to healthcare providers and provides temporal evidence of this over the clinical course of disease.

15.
Emerg Infect Dis ; 16(7): 1119-22, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20587184

RESUMO

Rhesus monkeys are protected from disease when a recombinant vesicular stomatitis virus-based vaccine is administered 20-30 min after infection with Marburg virus. We protected 5/6 monkeys when this vaccine was given 24 h after challenge; 2/6 animals were protected when the vaccine was administered 48 h postinfection.


Assuntos
Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/imunologia , Doenças dos Macacos/prevenção & controle , Vacinas Virais/imunologia , Animais , Macaca mulatta , Vacinas Sintéticas/imunologia , Vírus da Estomatite Vesicular Indiana/genética
16.
J Clin Microbiol ; 48(7): 2330-6, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20421440

RESUMO

Marburg virus (MARV) causes a severe hemorrhagic fever in humans with a high mortality rate. The rapid and accurate identification of the virus is required to appropriately provide infection control and outbreak management. Here, we developed and evaluated a one-step reverse transcription-loop-mediated isothermal amplification (RT-LAMP) assay for the rapid and simple detection of MARV. By combining two sets of primers specific for the Musoke and Ravn genetic lineages, a multiple RT-LAMP assay detected MARV strains of both lineages, and no cross-reactivity with other hemorrhagic fever viruses (Ebola virus and Lassa virus) was observed. The assay could detect 10(2) copies of the viral RNA per tube within 40 min by real-time monitoring of the turbidities of the reaction mixtures. The assay was further evaluated using viral RNA extracted from clinical specimens collected in the 2005 Marburg hemorrhagic fever outbreak in Angola and yielded positive results for samples containing MARV at greater than 10(4) 50% tissue culture infective doses/ml, exhibiting 78% (14 of 18 samples positive) consistency with the results of a reverse transcription-PCR assay carried out in the field laboratory. The results obtained by both agarose gel electrophoresis and naked-eye judgment indicated that the RT-LAMP assay developed in this study is an effective tool for the molecular detection of MARV. Furthermore, it seems suitable for use for field diagnostics or in laboratories in areas where MARV is endemic.


Assuntos
Marburgvirus , Técnicas de Diagnóstico Molecular/métodos , Técnicas de Amplificação de Ácido Nucleico/métodos , RNA Viral/análise , Virologia/métodos , Animais , Sequência de Bases , Chlorocebus aethiops , Humanos , Doença do Vírus de Marburg/diagnóstico , Marburgvirus/genética , Marburgvirus/isolamento & purificação , Dados de Sequência Molecular , Proteínas do Nucleocapsídeo , RNA Viral/isolamento & purificação , Transcrição Reversa , Ribonucleoproteínas/genética , Sensibilidade e Especificidade , Alinhamento de Sequência , Especificidade da Espécie , Células Vero , Proteínas Virais/genética
17.
J Virol ; 83(14): 7296-304, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19386702

RESUMO

The filoviruses Marburg virus and Ebola virus cause severe hemorrhagic fever with high mortality in humans and nonhuman primates. Among the most promising filovirus vaccines under development is a system based on recombinant vesicular stomatitis virus (VSV) that expresses a single filovirus glycoprotein (GP) in place of the VSV glycoprotein (G). Here, we performed a proof-of-concept study in order to determine the potential of having one single-injection vaccine capable of protecting nonhuman primates against Sudan ebolavirus (SEBOV), Zaire ebolavirus (ZEBOV), Cote d'Ivoire ebolavirus (CIEBOV), and Marburgvirus (MARV). In this study, 11 cynomolgus monkeys were vaccinated with a blended vaccine consisting of equal parts of the vaccine vectors VSVDeltaG/SEBOVGP, VSVDeltaG/ZEBOVGP, and VSVDeltaG/MARVGP. Four weeks later, three of these animals were challenged with MARV, three with CIEBOV, three with ZEBOV, and two with SEBOV. Three control animals were vaccinated with VSV vectors encoding a nonfilovirus GP and challenged with SEBOV, ZEBOV, and MARV, respectively, and five unvaccinated control animals were challenged with CIEBOV. Importantly, none of the macaques vaccinated with the blended vaccine succumbed to a filovirus challenge. As expected, an experimental control animal vaccinated with VSVDeltaG/ZEBOVGP and challenged with SEBOV succumbed, as did the positive controls challenged with SEBOV, ZEBOV, and MARV, respectively. All five control animals challenged with CIEBOV became severely ill, and three of the animals succumbed on days 12, 12, and 14, respectively. The two animals that survived CIEBOV infection were protected from subsequent challenge with either SEBOV or ZEBOV, suggesting that immunity to CIEBOV may be protective against other species of Ebola virus. In conclusion, we developed an immunization scheme based on a single-injection vaccine that protects nonhuman primates against lethal challenge with representative strains of all human pathogenic filovirus species.


Assuntos
Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/imunologia , Primatas/imunologia , Vacinas Virais/administração & dosagem , Animais , Anticorpos Antivirais/sangue , Modelos Animais de Doenças , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Humanos , Imunização , Imunoglobulina G/sangue , Macaca fascicularis , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/virologia , Primatas/virologia , Distribuição Aleatória , Vacinas Virais/imunologia
18.
PLoS Pathog ; 4(11): e1000225, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19043556

RESUMO

Ebola virus (EBOV) is a significant human pathogen that presents a public health concern as an emerging/re-emerging virus and as a potential biological weapon. Substantial progress has been made over the last decade in developing candidate preventive vaccines that can protect nonhuman primates against EBOV. Among these prospects, a vaccine based on recombinant vesicular stomatitis virus (VSV) is particularly robust, as it can also confer protection when administered as a postexposure treatment. A concern that has been raised regarding the replication-competent VSV vectors that express EBOV glycoproteins is how these vectors would be tolerated by individuals with altered or compromised immune systems such as patients infected with HIV. This is especially important as all EBOV outbreaks to date have occurred in areas of Central and Western Africa with high HIV incidence rates in the population. In order to address this concern, we evaluated the safety of the recombinant VSV vector expressing the Zaire ebolavirus glycoprotein (VSVDeltaG/ZEBOVGP) in six rhesus macaques infected with simian-human immunodeficiency virus (SHIV). All six animals showed no evidence of illness associated with the VSVDeltaG/ZEBOVGP vaccine, suggesting that this vaccine may be safe in immunocompromised populations. While one goal of the study was to evaluate the safety of the candidate vaccine platform, it was also of interest to determine if altered immune status would affect vaccine efficacy. The vaccine protected 4 of 6 SHIV-infected macaques from death following ZEBOV challenge. Evaluation of CD4+ T cells in all animals showed that the animals that succumbed to lethal ZEBOV challenge had the lowest CD4+ counts, suggesting that CD4+ T cells may play a role in mediating protection against ZEBOV.


Assuntos
Vacinas contra Ebola/farmacologia , Hospedeiro Imunocomprometido , Estomatite Vesicular , Animais , Linfócitos T CD4-Positivos , Avaliação Pré-Clínica de Medicamentos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Vacinas contra Ebola/administração & dosagem , Vetores Genéticos , Macaca mulatta , Primatas , Síndrome de Imunodeficiência Adquirida dos Símios/terapia , Resultado do Tratamento , Proteínas Virais
19.
PLoS Pathog ; 3(1): e2, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17238284

RESUMO

Ebola viruses are highly lethal human pathogens that have received considerable attention in recent years due to an increasing re-emergence in Central Africa and a potential for use as a biological weapon. There is no vaccine or treatment licensed for human use. In the past, however, important advances have been made in developing preventive vaccines that are protective in animal models. In this regard, we showed that a single injection of a live-attenuated recombinant vesicular stomatitis virus vector expressing the Ebola virus glycoprotein completely protected rodents and nonhuman primates from lethal Ebola challenge. In contrast, progress in developing therapeutic interventions against Ebola virus infections has been much slower and there is clearly an urgent need to develop effective post-exposure strategies to respond to future outbreaks and acts of bioterrorism, as well as to treat laboratory exposures. Here we tested the efficacy of the vesicular stomatitis virus-based Ebola vaccine vector in post-exposure treatment in three relevant animal models. In the guinea pig and mouse models it was possible to protect 50% and 100% of the animals, respectively, following treatment as late as 24 h after lethal challenge. More important, four out of eight rhesus macaques were protected if treated 20 to 30 min following an otherwise uniformly lethal infection. Currently, this approach provides the most effective post-exposure treatment strategy for Ebola infections and is particularly suited for use in accidentally exposed individuals and in the control of secondary transmission during naturally occurring outbreaks or deliberate release.


Assuntos
Vacinas contra Ebola/uso terapêutico , Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/terapia , Animais , Surtos de Doenças , Cobaias , Humanos , Macaca mulatta , Camundongos , Dados de Sequência Molecular , Resultado do Tratamento
20.
mBio ; 10(5)2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31641086

RESUMO

The 1918 influenza virus, subtype H1N1, was the causative agent of the most devastating pandemic in the history of infectious diseases. In vitro studies have confirmed that extreme virulence is an inherent property of this virus. Here, we utilized the macaque model for evaluating the efficacy of oseltamivir phosphate against the fully reconstructed 1918 influenza virus in a highly susceptible and relevant disease model. Our findings demonstrate that oseltamivir phosphate is effective in preventing severe disease in macaques but vulnerable to virus escape through emergence of resistant mutants, especially if given in a treatment regimen. Nevertheless, we conclude that oseltamivir would be highly beneficial to reduce the morbidity and mortality rates caused by a highly pathogenic influenza virus although it would be predicted that resistance would likely emerge with sustained use of the drug.IMPORTANCE Oseltamivir phosphate is used as a first line of defense in the event of an influenza pandemic prior to vaccine administration. Treatment failure through selection and replication of drug-resistant viruses is a known complication in the field and was also demonstrated in our study with spread of resistant 1918 influenza virus in multiple respiratory tissues. This emphasizes the importance of early treatment and the possibility that noncompliance may exacerbate treatment effectiveness. It also demonstrates the importance of implementing combination therapy and vaccination strategies as soon as possible in a pandemic situation.


Assuntos
Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/patogenicidade , Infecções por Orthomyxoviridae/tratamento farmacológico , Oseltamivir/uso terapêutico , Animais , Macaca , Infecções por Orthomyxoviridae/virologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa