Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Nature ; 531(7594): 381-5, 2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26934220

RESUMO

The most recent Ebola virus outbreak in West Africa, which was unprecedented in the number of cases and fatalities, geographic distribution, and number of nations affected, highlights the need for safe, effective, and readily available antiviral agents for treatment and prevention of acute Ebola virus (EBOV) disease (EVD) or sequelae. No antiviral therapeutics have yet received regulatory approval or demonstrated clinical efficacy. Here we report the discovery of a novel small molecule GS-5734, a monophosphoramidate prodrug of an adenosine analogue, with antiviral activity against EBOV. GS-5734 exhibits antiviral activity against multiple variants of EBOV and other filoviruses in cell-based assays. The pharmacologically active nucleoside triphosphate (NTP) is efficiently formed in multiple human cell types incubated with GS-5734 in vitro, and the NTP acts as an alternative substrate and RNA-chain terminator in primer-extension assays using a surrogate respiratory syncytial virus RNA polymerase. Intravenous administration of GS-5734 to nonhuman primates resulted in persistent NTP levels in peripheral blood mononuclear cells (half-life, 14 h) and distribution to sanctuary sites for viral replication including testes, eyes, and brain. In a rhesus monkey model of EVD, once-daily intravenous administration of 10 mg kg(-1) GS-5734 for 12 days resulted in profound suppression of EBOV replication and protected 100% of EBOV-infected animals against lethal disease, ameliorating clinical disease signs and pathophysiological markers, even when treatments were initiated three days after virus exposure when systemic viral RNA was detected in two out of six treated animals. These results show the first substantive post-exposure protection by a small-molecule antiviral compound against EBOV in nonhuman primates. The broad-spectrum antiviral activity of GS-5734 in vitro against other pathogenic RNA viruses, including filoviruses, arenaviruses, and coronaviruses, suggests the potential for wider medical use. GS-5734 is amenable to large-scale manufacturing, and clinical studies investigating the drug safety and pharmacokinetics are ongoing.


Assuntos
Alanina/análogos & derivados , Antivirais/uso terapêutico , Doença pelo Vírus Ebola/tratamento farmacológico , Macaca mulatta/virologia , Ribonucleotídeos/uso terapêutico , Monofosfato de Adenosina/análogos & derivados , Alanina/farmacocinética , Alanina/farmacologia , Alanina/uso terapêutico , Sequência de Aminoácidos , Animais , Antivirais/farmacocinética , Antivirais/farmacologia , Linhagem Celular Tumoral , Ebolavirus/efeitos dos fármacos , Feminino , Células HeLa , Doença pelo Vírus Ebola/prevenção & controle , Humanos , Masculino , Dados de Sequência Molecular , Especificidade de Órgãos , Pró-Fármacos/farmacocinética , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Ribonucleotídeos/farmacocinética , Ribonucleotídeos/farmacologia
2.
J Infect Dis ; 224(4): 632-642, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-33367826

RESUMO

BACKGROUND: Ebola virus disease (EVD) supportive care strategies are largely guided by retrospective observational research. This study investigated the effect of EVD supportive care algorithms on duration of survival in a controlled nonhuman primate (NHP) model. METHODS: Fourteen rhesus macaques were challenged intramuscularly with a target dose of Ebola virus (1000 plaque-forming units; Kikwit). NHPs were allocated to intensive care unit (ICU)-like algorithms (n = 7), intravenous fluids plus levofloxacin (n = 2), or a control group (n = 5). The primary outcome measure was duration of survival, and secondary outcomes included changes in clinical laboratory values. RESULTS: Duration of survival was not significantly different between the pooled ICU-like algorithm and control groups (8.2 vs 6.9 days of survival; hazard ratio; 0.50; P = .25). Norepinephrine was effective in transiently maintaining baseline blood pressure. NHPs treated with ICU-like algorithms had delayed onset of liver and kidney injury. CONCLUSIONS: While an obvious survival difference was not observed with ICU-like care, clinical observations from this model may aid in EVD supportive care NHP model refinement.


Assuntos
Cuidados Críticos , Doença pelo Vírus Ebola , Unidades de Terapia Intensiva , Animais , Modelos Animais de Doenças , Ebolavirus , Doença pelo Vírus Ebola/terapia , Macaca mulatta , Primatas , Estudos Retrospectivos
3.
J Infect Dis ; 222(11): 1894-1901, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-32479636

RESUMO

Marburg virus (MARV) is a filovirus with documented human case-fatality rates of up to 90%. Here, we evaluated the therapeutic efficacy of remdesivir (GS-5734) in nonhuman primates experimentally infected with MARV. Beginning 4 or 5 days post inoculation, cynomolgus macaques were treated once daily for 12 days with vehicle, 5 mg/kg remdesivir, or a 10-mg/kg loading dose followed by 5 mg/kg remdesivir. All vehicle-control animals died, whereas 83% of animals receiving a 10-mg/kg loading dose of remdesivir survived, as did 50% of animals receiving a 5-mg/kg remdesivir regimen. Remdesivir-treated animals exhibited improved clinical scores, lower plasma viral RNA, and improved markers of kidney function, liver function, and coagulopathy versus vehicle-control animals. The small molecule remdesivir showed therapeutic efficacy in this Marburg virus disease model with treatment initiation 5 days post inoculation, supporting further assessment of remdesivir for the treatment of Marburg virus disease in humans.


Assuntos
Antimetabólitos/uso terapêutico , Antivirais/uso terapêutico , Doença do Vírus de Marburg/tratamento farmacológico , Marburgvirus/efeitos dos fármacos , Doenças dos Macacos/tratamento farmacológico , Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Animais , Modelos Animais de Doenças , Feminino , Estimativa de Kaplan-Meier , Macaca fascicularis , Masculino , Doença do Vírus de Marburg/mortalidade , Doença do Vírus de Marburg/patologia , Doença do Vírus de Marburg/virologia , Doenças dos Macacos/mortalidade , Doenças dos Macacos/patologia , Doenças dos Macacos/virologia , RNA Viral
4.
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
5.
N Engl J Med ; 373(4): 339-48, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26200980

RESUMO

BACKGROUND: AVI-7288 is a phosphorodiamidate morpholino oligomer with positive charges that targets the viral messenger RNA that encodes Marburg virus (MARV) nucleoprotein. Its safety in humans is undetermined. METHODS: We assessed the efficacy of AVI-7288 in a series of studies involving a lethal challenge with MARV in nonhuman primates. The safety of AVI-7288 was evaluated in a randomized, multiple-ascending-dose study in which 40 healthy humans (8 humans per dose group) received 14 once-daily infusions of AVI-7288 (1 mg, 4 mg, 8 mg, 12 mg, or 16 mg per kilogram of body weight) or placebo, in a 3:1 ratio. We estimated the protective dose in humans by comparing pharmacokinetic variables in infected nonhuman primates, uninfected nonhuman primates, and uninfected humans. RESULTS: Survival in infected nonhuman primates was dose-dependent, with survival rates of 0%, 30%, 59%, 87%, 100%, and 100% among monkeys treated with 0 mg, 3.75 mg, 7.5 mg, 15 mg, 20 mg, and 30 mg of AVI-7288 per kilogram, respectively (P<0.001 with the use of the log-rank test for the comparison of survival across groups). No safety concern was identified at doses up to 16 mg per kilogram per day in humans. No serious adverse events were reported. Drug exposure (the area under the curve) was dose-dependent in both nonhuman primates and humans; drug clearance was independent of dose but was higher in nonhuman primates than in humans. The protective dose in humans was initially estimated, on the basis of exposure, to be 9.6 mg per kilogram per day (95% confidence interval, 6.6 to 12.5) for 14 days. Monte Carlo simulations supported a dose of 11 mg per kilogram per day to match the geometric mean protective exposure in nonhuman primates. CONCLUSIONS: This study shows that, on the basis of efficacy in nonhuman primates and pharmacokinetic data in humans, AVI-7288 has potential as postexposure prophylaxis for MARV infection in humans. (Funded by the Department of Defense; ClinicalTrials.gov number, NCT01566877.).


Assuntos
Antivirais/administração & dosagem , Doença do Vírus de Marburg/tratamento farmacológico , Marburgvirus , Morfolinos/administração & dosagem , Animais , Antivirais/efeitos adversos , Antivirais/farmacocinética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Estimativa de Kaplan-Meier , Macaca fascicularis , Doença do Vírus de Marburg/mortalidade , Marburgvirus/genética , Morfolinos/efeitos adversos , Morfolinos/farmacocinética , RNA Mensageiro , RNA Viral
6.
Emerg Infect Dis ; 23(8): 1316-1324, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28726603

RESUMO

The Ebola virus (EBOV) outbreak in West Africa during 2013-2016 demonstrated the need to improve Ebola virus disease (EVD) diagnostics and standards of care. This retrospective study compared laboratory values and clinical features of 3 nonhuman primate models of lethal EVD to assess associations with improved survival time. In addition, the study identified laboratory values useful as predictors of survival, surrogates for EBOV viral loads, and triggers for initiation of therapeutic interventions in these nonhuman primate models. Furthermore, the data support that, in nonhuman primates, the Makona strain of EBOV may be less virulent than the Kikwit strain of EBOV. The applicability of these findings as potential diagnostic and management tools for EVD in humans warrants further investigation.


Assuntos
Ebolavirus , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/virologia , Animais , Biomarcadores , Doença pelo Vírus Ebola/mortalidade , Doença pelo Vírus Ebola/transmissão , Humanos , Estimativa de Kaplan-Meier , Primatas , RNA Viral , Curva ROC , Estudos Retrospectivos , Carga Viral
7.
J Virol ; 88(2): 811-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24198421

RESUMO

Sin Nombre virus (SNV) is a rodent-borne hantavirus that causes hantavirus pulmonary syndrome (HPS) predominantly in North America. SNV infection of immunocompetent hamsters results in an asymptomatic infection; the only lethal disease model for a pathogenic hantavirus is Andes virus (ANDV) infection of Syrian hamsters. Efforts to create a lethal SNV disease model in hamsters by repeatedly passaging virus through the hamster have demonstrated increased dissemination of the virus but no signs of disease. In this study, we demonstrate that immunosuppression of hamsters through the administration of a combination of dexamethasone and cyclophosphamide, followed by infection with SNV, results in a vascular leak syndrome that accurately mimics both HPS disease in humans and ANDV infection of hamsters. Immunosuppressed hamsters infected with SNV have a mean number of days to death of 13 and display clinical signs associated with HPS, including pulmonary edema. Viral antigen was widely detectable throughout the pulmonary endothelium. Histologic analysis of lung sections showed marked inflammation and edema within the alveolar septa of SNV-infected hamsters, results which are similar to what is exhibited by hamsters infected with ANDV. Importantly, SNV-specific neutralizing polyclonal antibody administered 5 days after SNV infection conferred significant protection against disease. This experiment not only demonstrated that the disease was caused by SNV, it also demonstrated the utility of this animal model for testing candidate medical countermeasures. This is the first report of lethal disease caused by SNV in an adult small-animal model.


Assuntos
Modelos Animais de Doenças , Síndrome Pulmonar por Hantavirus/imunologia , Síndrome Pulmonar por Hantavirus/virologia , Mesocricetus , Vírus Sin Nombre/fisiologia , Animais , Anticorpos Antivirais/uso terapêutico , Cricetinae , Ciclofosfamida/administração & dosagem , Dexametasona/administração & dosagem , Feminino , Síndrome Pulmonar por Hantavirus/tratamento farmacológico , Síndrome Pulmonar por Hantavirus/mortalidade , Síndrome Pulmonar por Hantavirus/patologia , Humanos , Terapia de Imunossupressão , Imunossupressores/administração & dosagem
8.
Sci Rep ; 13(1): 3131, 2023 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-36823196

RESUMO

Remdesivir (GS-5734; VEKLURY) is a single diastereomer monophosphoramidate prodrug of an adenosine analog (GS-441524). Remdesivir is taken up by target cells and metabolized in multiple steps to form the active nucleoside triphosphate (GS-443902), which acts as a potent inhibitor of viral RNA-dependent RNA polymerases. Remdesivir and GS-441524 have antiviral activity against multiple RNA viruses. Here, we expand the evaluation of remdesivir's antiviral activity to members of the families Flaviviridae, Picornaviridae, Filoviridae, Orthomyxoviridae, and Hepadnaviridae. Using cell-based assays, we show that remdesivir can inhibit infection of flaviviruses (such as dengue 1-4, West Nile, yellow fever, Zika viruses), picornaviruses (such as enterovirus and rhinovirus), and filoviruses (such as various Ebola, Marburg, and Sudan virus isolates, including novel geographic isolates), but is ineffective or is significantly less effective against orthomyxoviruses (influenza A and B viruses), or hepadnaviruses B, D, and E. In addition, remdesivir shows no antagonistic effect when combined with favipiravir, another broadly acting antiviral nucleoside analog, and has minimal interaction with a panel of concomitant medications. Our data further support remdesivir as a broad-spectrum antiviral agent that has the potential to address multiple unmet medical needs, including those related to antiviral pandemic preparedness.


Assuntos
Filoviridae , Doença pelo Vírus Ebola , Infecção por Zika virus , Zika virus , Humanos , Antivirais/farmacologia , Antivirais/uso terapêutico , Monofosfato de Adenosina , Alanina , Doença pelo Vírus Ebola/tratamento farmacológico , Infecção por Zika virus/tratamento farmacológico
9.
Viruses ; 15(12)2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-38140576

RESUMO

Marburg virus (MARV) causes severe disease and high mortality in humans. The objective of this study was to characterize disease manifestations and pathogenesis in cynomolgus macaques exposed to MARV. The results of this natural history study may be used to identify features of MARV disease useful in defining the ideal treatment initiation time for subsequent evaluations of investigational therapeutics using this model. Twelve cynomolgus macaques were exposed to a target dose of 1000 plaque-forming units MARV by the intramuscular route, and six control animals were mock-exposed. The primary endpoint of this study was survival to Day 28 post-inoculation (PI). Anesthesia events were minimized with the use of central venous catheters for periodic blood collection, and temperature and activity were continuously monitored by telemetry. All mock-exposed animals remained healthy for the duration of the study. All 12 MARV-exposed animals (100%) became infected, developed illness, and succumbed on Days 8-10 PI. On Day 4 PI, 11 of the 12 MARV-exposed animals had statistically significant temperature elevations over baseline. Clinically observable signs of MARV disease first appeared on Day 5 PI, when 6 of the 12 animals exhibited reduced responsiveness. Ultimately, systemic inflammation, coagulopathy, and direct cytopathic effects of MARV all contributed to multiorgan dysfunction, organ failure, and death or euthanasia of all MARV-exposed animals. Manifestations of MARV disease, including fever, systemic viremia, lymphocytolysis, coagulopathy, and hepatocellular damage, could be used as triggers for initiation of treatment in future therapeutic efficacy studies.


Assuntos
Doença do Vírus de Marburg , Marburgvirus , Humanos , Animais , Macaca fascicularis , Viremia , Fígado
10.
J Virol ; 85(8): 3905-17, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21289122

RESUMO

Marburgvirus (MARV) infections are generally lethal in humans and nonhuman primates but require in vivo lethal mouse variant selection by the serial transfer (passage) of the nonlethal virus into naïve mice to propagate a lethal infection. The passage of progenitor (wild-type) MARV or Ravn virus (RAVV) from infected scid BALB/c mouse liver homogenates into immunocompetent BALB/c mice results in the selection of lethal mouse viruses from within the quasispecies sufficient to establish lethality in immunocompetent mice. Genomic analysis in conjunction with the passage history of each mutation detailed the altered primary and secondary structures of the viral genomic RNA throughout the process. Key findings included the following: (i) a VP40:D184N mutation previously identified in the lethal guinea pig MARV genome was the first mutation to occur during the passage of both the MARV and RAVV variants; (ii) there was biased hypermutagenesis in the RAVV variant genome; (iii) there were two identical mutations in lethal mouse MARV and RAVV variants, VP40:Y19H in the PPPY motif and VP40:D184N in a loop structure between the two VP40 domains; (iv) the passage of wild-type MARV and RAVV in mice resulted in the selection of viral variants from among the quasispecies with different genotypes than those of the wild-type viruses; and (v) a lethal mouse RAVV variant had different tissue tropisms distinct from those of its wild-type virus. These studies provide insights into how marburgviruses manipulate the host for enzymes, metabolites, translation regulators, and effectors of the innate immune response to serve as potential viral countermeasures.


Assuntos
Evolução Molecular , Genoma Viral , Doença do Vírus de Marburg/virologia , Marburgvirus/genética , Marburgvirus/patogenicidade , Doenças dos Roedores/virologia , Seleção Genética , Substituição de Aminoácidos/genética , Animais , Análise Mutacional de DNA , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mutação de Sentido Incorreto , RNA Viral/genética , Doenças dos Roedores/mortalidade , Inoculações Seriadas , Análise de Sobrevida , Tropismo Viral
11.
Sci Rep ; 11(1): 19458, 2021 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-34593911

RESUMO

Efficacious therapeutics for Ebola virus disease are in great demand. Ebola virus infections mediated by mucosal exposure, and aerosolization in particular, present a novel challenge due to nontypical massive early infection of respiratory lymphoid tissues. We performed a randomized and blinded study to compare outcomes from vehicle-treated and remdesivir-treated rhesus monkeys in a lethal model of infection resulting from aerosolized Ebola virus exposure. Remdesivir treatment initiated 4 days after exposure was associated with a significant survival benefit, significant reduction in serum viral titer, and improvements in clinical pathology biomarker levels and lung histology compared to vehicle treatment. These observations indicate that remdesivir may have value in countering aerosol-induced Ebola virus disease.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Doença pelo Vírus Ebola/tratamento farmacológico , Monofosfato de Adenosina/administração & dosagem , Monofosfato de Adenosina/farmacologia , Administração Intravenosa , Aerossóis , Alanina/administração & dosagem , Alanina/farmacologia , Animais , Antivirais/administração & dosagem , Modelos Animais de Doenças , Feminino , Doença pelo Vírus Ebola/sangue , Estimativa de Kaplan-Meier , Fígado/efeitos dos fármacos , Fígado/virologia , Pulmão/patologia , Pulmão/virologia , Linfonodos/efeitos dos fármacos , Linfonodos/patologia , Linfonodos/virologia , Macaca mulatta , Masculino , Distribuição Aleatória , Síndrome de Resposta Inflamatória Sistêmica/tratamento farmacológico , Síndrome de Resposta Inflamatória Sistêmica/virologia , Carga Viral/efeitos dos fármacos , Viremia/tratamento farmacológico
12.
Antimicrob Agents Chemother ; 54(5): 2152-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20211898

RESUMO

There exists an urgent need to develop licensed drugs and vaccines for the treatment or prevention of filovirus infections. FGI-103 is a low-molecular-weight compound that was discovered through an in vitro screening assay utilizing a variant of Zaire ebolavirus (ZEBOV) that expresses green fluorescent protein. In vitro analyses demonstrated that FGI-103 also exhibits antiviral activity against wild-type ZEBOV and Sudan ebolavirus, as well as Marburgvirus (MARV) strains Ci67 and Ravn. In vivo administration of FGI-103 as a single intraperitoneal dose of 10 mg/kg delivered 24 h after infection is sufficient to completely protect mice against a lethal challenge with a mouse-adapted strain of either ZEBOV or MARV-Ravn. In a murine model of ZEBOV infection, delivery of FGI-103 reduces viremia and the viral burden in kidney, liver, and spleen tissues and is associated with subdued and delayed proinflammatory cytokine responses and tissue pathology. Taken together, these results identify a promising antiviral therapeutic candidate for the treatment of filovirus infections.


Assuntos
Amidinas/farmacologia , Antivirais/farmacologia , Benzofuranos/farmacologia , Infecções por Filoviridae/tratamento farmacológico , Filoviridae/efeitos dos fármacos , Amidinas/química , Animais , Antivirais/química , Benzofuranos/química , Chlorocebus aethiops , Citocinas/metabolismo , Ebolavirus/efeitos dos fármacos , Ebolavirus/genética , Feminino , Filoviridae/genética , Infecções por Filoviridae/imunologia , Infecções por Filoviridae/patologia , Proteínas de Fluorescência Verde/genética , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/patologia , Células Hep G2 , Humanos , Rim/citologia , Fígado/patologia , Fígado/virologia , Masculino , Doença do Vírus de Marburg/tratamento farmacológico , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/patologia , Marburgvirus/efeitos dos fármacos , Marburgvirus/genética , Camundongos , Camundongos Endogâmicos C57BL , Peso Molecular , Bibliotecas de Moléculas Pequenas , Células Vero
13.
J Virol ; 83(13): 6404-15, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19369350

RESUMO

The lack of a mouse model has hampered an understanding of the pathogenesis and immunity of Marburg hemorrhagic fever (MHF), the disease caused by marburgvirus (MARV), and has created a bottleneck in the development of antiviral therapeutics. Primary isolates of the filoviruses, i.e., ebolavirus (EBOV) and MARV, are not lethal to immunocompetent adult mice. Previously, pathological, virologic, and immunologic evaluation of a mouse-adapted EBOV, developed by sequential passages in suckling mice, identified many similarities between this model and EBOV infections in nonhuman primates. We recently demonstrated that serially passaging virus recovered from the liver homogenates of MARV-infected immunodeficient (SCID) mice was highly successful in reducing the time to death in these mice from 50 to 70 days to 7 to 10 days after challenge with the isolate MARV-Ci67, -Musoke, or -Ravn. In this study, we extended our findings to show that further sequential passages of MARV-Ravn in immunocompetent mice caused the MARV to kill BALB/c mice. Serial sampling studies to characterize the pathology of mouse-adapted MARV-Ravn revealed that this model is similar to the guinea pig and nonhuman primate MHF models. Infection of BALB/c mice with mouse-adapted MARV-Ravn caused uncontrolled viremia and high viral titers in the liver, spleen, lymph node, and other organs; profound lymphopenia; destruction of lymphocytes within the spleen and lymph nodes; and marked liver damage and thrombocytopenia. Sequencing the mouse-adapted MARV-Ravn strain revealed differences in 16 predicted amino acids from the progenitor virus, although the exact changes required for adaptation are unclear at this time. This mouse-adapted MARV strain can now be used to develop and evaluate novel vaccines and therapeutics and may also help to provide a better understanding of the virulence factors associated with MARV.


Assuntos
Modelos Animais de Doenças , Doença do Vírus de Marburg/virologia , Marburgvirus/patogenicidade , Animais , Chlorocebus aethiops , Feminino , Fígado/patologia , Fígado/virologia , Masculino , Marburgvirus/genética , Camundongos , Camundongos Endogâmicos BALB C , RNA Viral/genética , Análise de Sequência de RNA , Inoculações Seriadas , Baço/patologia , Baço/virologia , Células Vero
14.
Viruses ; 12(1)2020 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-31941095

RESUMO

Recent Ebola virus (EBOV) outbreaks in West Africa and the Democratic Republic of the Congo have highlighted the urgent need for approval of medical countermeasures for treatment and prevention of EBOV disease (EVD). Until recently, when successes were achieved in characterizing the efficacy of multiple experimental EVD therapeutics in humans, the only feasible way to obtain data regarding potential clinical benefits of candidate therapeutics was by conducting well-controlled animal studies. Nonclinical studies are likely to continue to be important tools for screening and development of new candidates with improved pharmacological properties. Here, we describe a natural history study to characterize the time course and order of progression of the disease manifestations of EVD in rhesus monkeys. In 12 rhesus monkeys exposed by the intramuscular route to 1000 plaque-forming units of EBOV, multiple endpoints were monitored for 28 days following exposure. The disease progressed rapidly with mortality events occurring 7-10 days after exposure. Key disease manifestations observed consistently across the infected animals included, but were not limited to, viremia, fever, systemic inflammation, coagulopathy, lymphocytolysis, renal tubular necrosis with mineralization, and hepatocellular degeneration and necrosis.


Assuntos
Modelos Animais de Doenças , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/fisiopatologia , Macaca mulatta/virologia , Animais , Progressão da Doença , Feminino , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/mortalidade , Injeções Intramusculares , Masculino
15.
Antiviral Res ; 171: 104592, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31473342

RESUMO

Filoviridae currently includes five official and one proposed genera. Genus Ebolavirus includes five established and one proposed ebolavirus species for Bombali virus (BOMV), Bundibugyo virus (BDBV), Ebola virus (EBOV), Reston virus (RESTV), Sudan virus (SUDV) and Taï Forest virus (TAFV), and genus Marburgvirus includes a single species for Marburg virus (MARV) and Ravn virus (RAVV). Ebola virus (EBOV) has emerged as a significant public health concern since the 2013-2016 Ebola Virus Disease outbreak in Western Africa. Currently, there are no therapeutics approved and the need for Ebola-specific therapeutics remains a gap. In search for anti-Ebola therapies we tested the idea of using inhibitory properties of peptides corresponding to the C-terminal heptad-repeat (HR2) domains of class I fusion proteins against EBOV infection. The fusion protein GP2 of EBOV belongs to class I, suggesting that a similar strategy to HIV may be applied to inhibit EBOV infection. The serum half-life of peptides was expanded by cholesterol conjugation to allow daily dosing. The peptides were further constrained to stabilize a helical structure to increase the potency of inhibition. The EC50s of lead peptides were in low micromolar range, as determined by a high-content imaging test of EBOV-infected cells. Lead peptides were tested in an EBOV lethal mouse model and efficacy of the peptides were determined following twice-daily administration of peptides for 9 days. The most potent peptide was able to protect mice from lethal challenge of mouse-adapted Ebola virus. These data show that engineered peptides coupled with cholesterol can inhibit viral production, protect mice against lethal EBOV infection, and may be used to build novel therapeutics against EBOV.


Assuntos
Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Marburgvirus/efeitos dos fármacos , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Antivirais/química , Linhagem Celular , Colesterol/química , Modelos Animais de Doenças , Doença pelo Vírus Ebola/virologia , Doença do Vírus de Marburg/virologia , Camundongos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Peptídeos/química , Conformação Proteica , Relação Estrutura-Atividade
16.
Antiviral Res ; 151: 50-54, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29289664

RESUMO

During the 2013-2016 Ebola virus (EBOV) outbreak in West Africa, our team at USAMRIID evaluated the antiviral activity of a number of compounds, including favipiravir (T-705), in vitro and in mouse and nonhuman primate (NHP) models of Ebola virus disease. In this short communication, we present our findings for favipiravir in cell culture and in mice, while an accompanying paper presents the results of NHP studies. We confirmed previous reports that favipiravir has anti-EBOV activity in mice. Additionally, we found that the active form of favipiravir is generated in mice in tissues relevant for the pathogenesis of EBOV infection. Finally, we observed that protection can be achieved in mice down to 8 mg/kg/day, which is lower than the dosing regimens previously reported. An accompanying paper reports the results of treating nonhuman primates infected with EBOV or with Marburg virus with oral or intravenous favipiravir.


Assuntos
Amidas/farmacologia , Amidas/uso terapêutico , Ebolavirus/efeitos dos fármacos , Doença pelo Vírus Ebola/tratamento farmacológico , Pirazinas/farmacologia , Pirazinas/uso terapêutico , Amidas/metabolismo , Animais , Antivirais/metabolismo , Antivirais/farmacologia , Antivirais/uso terapêutico , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citoplasma/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Marburgvirus/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Pirazinas/metabolismo , Análise de Sobrevida , Replicação Viral/efeitos dos fármacos
17.
Antiviral Res ; 151: 97-104, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29289666

RESUMO

Favipiravir is a broad-spectrum antiviral agent that has demonstrated efficacy against Ebola virus (EBOV) in rodents. However, there are no published reports of favipiravir efficacy for filovirus infection of nonhuman primates (NHPs). Here we evaluated the pharmacokinetic profile of favipiravir in NHPs, as well as in vivo efficacy against two filoviruses, EBOV and Marburg virus (MARV). While no survival benefit was observed in two studies employing once- or twice-daily oral dosing of favipiravir during EBOV infection of NHPs, an antiviral effect was observed in terms of extended time-to-death and reduced levels of viral RNA. However, oral dosing in biosafety level-4 (BSL-4) presents logistical and technical challenges, and repeated anesthesia events may potentially worsen survival outcome in animals. For the third study of treatment of MARV infection, we therefore made use of catheters, jackets, and tethers for intravenous (IV) dosing and blood collection, which minimized the requirement for repeated anesthesia events. When MARV infection was treated with IV favipiravir, five of six animals (83%) survived infection, while all untreated NHPs succumbed. An accompanying report presents the results of favipiravir treatment of EBOV infection in mice.


Assuntos
Amidas/administração & dosagem , Amidas/farmacologia , Ebolavirus/efeitos dos fármacos , Doença pelo Vírus Ebola/tratamento farmacológico , Doença do Vírus de Marburg/tratamento farmacológico , Marburgvirus/efeitos dos fármacos , Pirazinas/administração & dosagem , Pirazinas/farmacologia , Animais , Antivirais/administração & dosagem , Antivirais/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Doença pelo Vírus Ebola/patologia , Doença pelo Vírus Ebola/virologia , Masculino , Doença do Vírus de Marburg/patologia , Doença do Vírus de Marburg/virologia , Primatas , RNA Viral/sangue , Análise de Sobrevida , Carga Viral/efeitos dos fármacos
18.
Antiviral Res ; 73(2): 118-25, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16987555

RESUMO

The fully glycosylated human omega interferon produced from CHO-SS cells (glycosylated IFN-omega) has been shown to be well-tolerated in man and to induce a sustained virologic response in patients infected with hepatitis C virus (HCV). We examined the antiviral activity of glycosylated IFN-omega and various human IFNs (IFN-alpha, -beta, -gamma and non-glycosylated bacterial (Escherichia coli) recombinant IFN-omega (non-glycosylated IFN-omega)) against HCV RNA replicons and several viruses related to HCV. Since none of the IFNs displayed cytotoxicity we compared their activities based on the effective concentration of the IFN that inhibited virus growth by 50% (EC50). Glycosylated IFN-omega was found to be the most potent antiviral agent of all the IFNs tested against bovine viral diarrhea virus (BVDV), yellow fever virus and West Nile virus. With HCV RNA replicons, non-glycosylated IFN-omega was comparable in activity to IFN-alpha while glycosylated IFN-omega was markedly more potent, indicating that glycosylation has an important effect on its activity. Drug combination analysis of glycosylated IFN-omega+ribavirin (RBV) in BVDV showed a synergy of antiviral effects similar to IFN-alpha+RBV, as well as a unique antagonism of RBV cytotoxic effects by glycosylated IFN-omega. Transcription factor (TF) profiling indicated that IFN-alpha or glycosylated IFN-omega treatment upregulated the same 17 TFs. IFN-alpha and glycosylated IFN-omega also upregulated 9 and 40 additional unique TFs, respectively. The differences in the expression of these TFs were modest, but statistically significantly different for eight of the TFs that were upregulated exclusively by glycosylated IFN-omega. The activation of these additional TFs by glycosylated IFN-omega might contribute to its high potency.


Assuntos
Antivirais/farmacologia , Hepacivirus/genética , Interferon Tipo I/farmacologia , Interferons/farmacologia , Replicon/efeitos dos fármacos , Animais , Antivirais/imunologia , Células CHO , Bovinos , Chlorocebus aethiops , Cricetinae , Cricetulus , Combinação de Medicamentos , Humanos , Interferon Tipo I/imunologia , Interferons/imunologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Replicon/genética , Fatores de Transcrição/genética , Células Vero
19.
Antiviral Res ; 138: 22-31, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27908828

RESUMO

Iminosugars are host-directed antivirals with broad-spectrum activity. The iminosugar, N-butyl-deoxynojirimycin (NB-DNJ or Miglustat®), is used in humans for treatment of Gaucher's disease and has mild antiviral properties. More potent analogs of NB-DNJ have been generated and have demonstrated activity against a variety of viruses including flaviviruses, influenza, herpesviruses and filoviruses. In the current study, a panel of analogs based on NB-DNJ was analyzed for activity against Ebola (EBOV) and Marburg viruses (MARV). The antiviral activity of NB-DNJ (UV-1), UV-2, UV-3, UV-4 and UV-5 against both EBOV and MARV was demonstrated in Vero cells. Subsequent studies to examine the activity of UV-4 and UV-5 using rodent models of EBOV and MARV were performed. In vivo efficacy studies provided inconsistent data following treatment with iminosugars using filovirus mouse models. A tolerability study in nonhuman primates demonstrated that UV-4 could be administered at much higher dose levels than rodents. Since UV-4 was active in vitro, had been demonstrated to be active against influenza and dengue in vivo, and was being tested in a Phase 1 clinical trial, a small proof-of-concept nonhuman primate trial was performed to determine whether this antiviral candidate could provide clinical benefit to EBOV-infected individuals. Administration of UV-4B did not provide a clinical or survival benefit to macaques infected with EBOV-Makona; however, dosing of animals was not optimal in this study. Efficacy may be improved by thrice daily dosing (e.g. by nasogastric tube feeding) to match the efficacious dosing regimens demonstrated against dengue and influenza viruses.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , Ebolavirus/efeitos dos fármacos , Imino Açúcares/farmacologia , Imino Açúcares/uso terapêutico , Marburgvirus/efeitos dos fármacos , 1-Desoxinojirimicina/administração & dosagem , 1-Desoxinojirimicina/agonistas , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacologia , 1-Desoxinojirimicina/uso terapêutico , Animais , Antivirais/administração & dosagem , Antivirais/química , Chlorocebus aethiops , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Imino Açúcares/administração & dosagem , Imino Açúcares/química , Macaca , Camundongos , Modelos Animais , Células Vero
20.
Viruses ; 8(4): 94, 2016 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-27070636

RESUMO

Marburg virus (MARV) was the first filovirus to be identified following an outbreak of viral hemorrhagic fever disease in Marburg, Germany in 1967. Due to several factors inherent to filoviruses, they are considered a potential bioweapon that could be disseminated via an aerosol route. Previous studies demonstrated that MARV virus-like particles (VLPs) containing the glycoprotein (GP), matrix protein VP40 and nucleoprotein (NP) generated using a baculovirus/insect cell expression system could protect macaques from subcutaneous (SQ) challenge with multiple species of marburgviruses. In the current study, the protective efficacy of the MARV VLPs in conjunction with two different adjuvants: QS-21, a saponin derivative, and poly I:C against homologous aerosol challenge was assessed in cynomolgus macaques. Antibody responses against the GP antigen were equivalent in all groups receiving MARV VLPs irrespective of the adjuvant; adjuvant only-vaccinated macaques did not demonstrate appreciable antibody responses. All macaques were subsequently challenged with lethal doses of MARV via aerosol or SQ as a positive control. All MARV VLP-vaccinated macaques survived either aerosol or SQ challenge while animals administered adjuvant only exhibited clinical signs and lesions consistent with MARV disease and were euthanized after meeting the predetermined criteria. Therefore, MARV VLPs induce IgG antibodies recognizing MARV GP and VP40 and protect cynomolgus macaques from an otherwise lethal aerosol exposure with MARV.


Assuntos
Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/imunologia , Vacinação , Vacinas de Partículas Semelhantes a Vírus/imunologia , Animais , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Imunoglobulina G/imunologia , Fígado/imunologia , Fígado/metabolismo , Fígado/patologia , Macaca fascicularis , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/virologia , Baço/imunologia , Baço/metabolismo , Baço/patologia , Fatores de Tempo , Resultado do Tratamento , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Viremia/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA