Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Infect Dis ; 218(suppl_5): S592-S596, 2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-30016444

RESUMO

At the onset of the 2013-2016 epidemic of Ebola virus disease (EVD), no vaccine or antiviral medication was approved for treatment. Therefore, considerable efforts were directed towards the concept of drug repurposing or repositioning. Amiodarone, an approved multi-ion channel blocker for the treatment of cardiac arrhythmia, was reported to inhibit filovirus entry in vitro. Compassionate use of amiodarone in EVD patients indicated a possible survival benefit. In support of further clinical testing, we confirmed anti-Ebola virus activity of amiodarone in different cell types. Despite promising in vitro results, amiodarone failed to protect guinea pigs from a lethal dose of Ebola virus.


Assuntos
Amiodarona/farmacologia , Ebolavirus/efeitos dos fármacos , Amiodarona/farmacocinética , Amiodarona/uso terapêutico , Animais , Chlorocebus aethiops , Feminino , Cobaias , Doença pelo Vírus Ebola/tratamento farmacológico , Masculino , Células Vero
2.
J Infect Dis ; 218(suppl_5): S672-S678, 2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-29939303

RESUMO

Background: A need to develop therapeutics to treat Ebola virus disease patients in remote and resource-challenged settings remains in the wake of the 2013-2016 epidemic in West Africa. Toward this goal, we screened drugs under consideration as treatment options and other drugs of interest, most being small molecules approved by the Food and Drug Administration. Drugs demonstrating in vitro antiviral activity were advanced for evaluation in combinations because of advantages often provided by drug cocktails. Methods: Drugs were screened for blockade of Ebola virus infection in cultured cells. Twelve drugs were tested in all (78 pair-wise) combinations, and 3 were tested in a subset of combinations. Results: Multiple synergistic drug pairs emerged, with the majority comprising 2 entry inhibitors. For the pairs of entry inhibitors studied, synergy was demonstrated at the level of virus entry into host cells. Highly synergistic pairs included aripiprazole/piperacetazine, sertraline/toremifene, sertraline/bepridil, and amodiaquine/clomiphene. Conclusions: Our study shows the feasibility of identifying pairs of approved drugs that synergistically block Ebola virus infection in cell cultures. We discuss our findings in terms of the theoretic ability of these or alternate combinations to reach therapeutic levels. Future research will assess selected combinations in small-animal models of Ebola virus disease.


Assuntos
Antivirais/administração & dosagem , Ebolavirus/efeitos dos fármacos , Animais , Antivirais/uso terapêutico , Chlorocebus aethiops , Aprovação de Drogas , Sinergismo Farmacológico , Quimioterapia Combinada , Células Vero , Vírion/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos
3.
J Infect Dis ; 215(9): 1416-1420, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28368541

RESUMO

Previous studies have demonstrated little efficacy of interferons (IFNs) in animal models of Ebola virus disease. However, these studies were limited to a small number of type I IFNs and, during the most recent outbreak of Ebola virus, questions regarding the suitability of the animal models to evaluate IFNs were raised. To address the potential that anti-Ebola virus activity was overlooked, type I and type II IFNs (α-2a, α-2b, -ß, -γ, and -universal) were tested in a variety of cell types (Vero E6, Huh 7 cells, and human macrophages). IFNs are weak inhibitors of Ebola virus Makona in these cell lines.


Assuntos
Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Interferon beta/farmacologia , Interferon gama/farmacologia , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Sinergismo Farmacológico , Humanos , Células Vero
4.
Antimicrob Agents Chemother ; 59(2): 1088-99, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25487801

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) is a lineage C betacoronavirus, and infections with this virus can result in acute respiratory syndrome with renal failure. Globally, MERS-CoV has been responsible for 877 laboratory-confirmed infections, including 317 deaths, since September 2012. As there is a paucity of information regarding the molecular pathogenesis associated with this virus or the identities of novel antiviral drug targets, we performed temporal kinome analysis on human hepatocytes infected with the Erasmus isolate of MERS-CoV with peptide kinome arrays. bioinformatics analysis of our kinome data, including pathway overrepresentation analysis (ORA) and functional network analysis, suggested that extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) and phosphoinositol 3-kinase (PI3K)/serine-threonine kinase (AKT)/mammalian target of rapamycin (mTOR) signaling responses were specifically modulated in response to MERS-CoV infection in vitro throughout the course of infection. The overrepresentation of specific intermediates within these pathways determined by pathway and functional network analysis of our kinome data correlated with similar patterns of phosphorylation determined through Western blot array analysis. In addition, analysis of the effects of specific kinase inhibitors on MERS-CoV infection in tissue culture models confirmed these cellular response observations. Further, we have demonstrated that a subset of licensed kinase inhibitors targeting the ERK/MAPK and PI3K/AKT/mTOR pathways significantly inhibited MERS-CoV replication in vitro whether they were added before or after viral infection. Taken together, our data suggest that ERK/MAPK and PI3K/AKT/mTOR signaling responses play important roles in MERS-CoV infection and may represent novel drug targets for therapeutic intervention strategies.


Assuntos
Infecções por Coronavirus/metabolismo , Hepatócitos/metabolismo , Hepatócitos/virologia , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Western Blotting , Linhagem Celular , Biologia Computacional , Infecções por Coronavirus/virologia , Ensaio de Imunoadsorção Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Fosforilação , Transdução de Sinais/fisiologia
5.
Antimicrob Agents Chemother ; 58(8): 4885-93, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24841273

RESUMO

Outbreaks of emerging infections present health professionals with the unique challenge of trying to select appropriate pharmacologic treatments in the clinic with little time available for drug testing and development. Typically, clinicians are left with general supportive care and often untested convalescent-phase plasma as available treatment options. Repurposing of approved pharmaceutical drugs for new indications presents an attractive alternative to clinicians, researchers, public health agencies, drug developers, and funding agencies. Given the development times and manufacturing requirements for new products, repurposing of existing drugs is likely the only solution for outbreaks due to emerging viruses. In the studies described here, a library of 290 compounds was screened for antiviral activity against Middle East respiratory syndrome coronavirus (MERS-CoV) and severe acute respiratory syndrome coronavirus (SARS-CoV). Selection of compounds for inclusion in the library was dependent on current or previous FDA approval or advanced clinical development. Some drugs that had a well-defined cellular pathway as target were included. In total, 27 compounds with activity against both MERS-CoV and SARS-CoV were identified. The compounds belong to 13 different classes of pharmaceuticals, including inhibitors of estrogen receptors used for cancer treatment and inhibitors of dopamine receptor used as antipsychotics. The drugs identified in these screens provide new targets for in vivo studies as well as incorporation into ongoing clinical studies.


Assuntos
Antivirais/farmacologia , Reposicionamento de Medicamentos , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Antipsicóticos/farmacologia , Chlorocebus aethiops , Aprovação de Drogas , Antagonistas de Estrogênios/farmacologia , Ensaios de Triagem em Larga Escala , Humanos , Concentração Inibidora 50 , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Células Vero , Replicação Viral/efeitos dos fármacos
6.
J Gen Virol ; 95(Pt 3): 571-577, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24323636

RESUMO

The Middle East respiratory syndrome coronavirus (MERS-CoV) presents a novel emerging threat to public health worldwide. Several treatments for infected individuals have been suggested including IFN, ribavirin and passive immunotherapy with convalescent plasma. Administration of IFN-α2b and ribavirin has improved outcomes of MERS-CoV infection in rhesus macaques when administered within 8 h post-challenge. However, detailed and systematic evidence on the activity of other clinically available drugs is limited. Here we compared the susceptibility of MERS-CoV with different IFN products (IFN-α2b, IFN-γ, IFN-universal, IFN-α2a and IFN-ß), as well as with two antivirals, ribavirin and mycophenolic acid (MPA), against MERS-CoV (Hu/Jordan-N3/2012) in vitro. Of all the IFNs tested, IFN-ß showed the strongst inhibition of MERS-CoV in vitro, with an IC50 of 1.37 U ml(-1), 41 times lower than the previously reported IC50 (56.08 U ml(-1)) of IFN-α2b. IFN-ß inhibition was confirmed in the virus yield reduction assay, with an IC90 of 38.8 U ml(-1). Ribavirin did not inhibit viral replication in vitro at a dose that would be applicable to current treatment protocols in humans. In contrast, MPA showed strong inhibition, with an IC50 of 2.87 µM. This drug has not been previously tested against MERS-CoV and may provide an alternative to ribavirin for treatment of MERS-CoV. In conclusion, IFN-ß, MPA or a combination of the two may be beneficial in the treatment of MERS-CoV or as a post-exposure intervention in high-risk patients with known exposures to MERS-CoV.


Assuntos
Infecções por Coronaviridae/virologia , Coronaviridae/efeitos dos fármacos , Interferons/farmacologia , Ácido Micofenólico/farmacologia , Animais , Linhagem Celular , Chlorocebus aethiops , Coronaviridae/fisiologia , Infecções por Coronaviridae/tratamento farmacológico , Humanos , Células Vero , Replicação Viral/efeitos dos fármacos
7.
PLoS One ; 13(3): e0194868, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29566060

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) presents an emerging threat to public health worldwide by causing severe respiratory disease in humans with high virulence and case fatality rate (about 35%) since 2012. Little is known about the pathogenesis and innate antiviral response in primary human monocyte-derived macrophages (MDMs) and dendritic cells (MDDCs) upon MERS-CoV infection. In this study, we assessed MERS-CoV replication as well as induction of inflammatory cytokines and chemokines in MDMs and immature and mature MDDCs. Immature MDDCs and MDMs were permissive for MERS-CoV infection, while mature MDDCs were not, with stimulation of proinflammatory cytokine and chemokine upregulation in MDMs, but not in MDDCs. To further evaluate the antiviral activity of well-defined drugs in primary antigen presenting cells (APCs), three compounds (chloroquine, chlorpromazine and toremifine), each with broad-spectrum antiviral activity in immortalized cell lines, were evaluated in MDMs and MDDCs to determine their antiviral effect on MERS-CoV infection. While chloroquine was not active in these primary cells, chlorpromazine showed strong anti-MERS-CoV activity, but it was associated with high cytotoxicity narrowing the potential window for drug utilization. Unlike in established cells, toremifene had marginal activity when tested in antigen presenting cells, with high apparent cytotoxicity, also limiting its potential as a therapeutic option. These results demonstrate the value of testing drugs in primary cells, in addition to established cell lines, before initiating preclinical or clinical studies for MERS treatment and the importance of carefully assessing cytotoxicity in drug screen assays. Furthermore, these studies also highlight the role of APCs in stimulating a robust protective immune response to MERS-CoV infection.


Assuntos
Células Apresentadoras de Antígenos/efeitos dos fármacos , Antivirais/uso terapêutico , Infecções por Coronavirus/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Coronavírus da Síndrome Respiratória do Oriente Médio , Animais , Células Apresentadoras de Antígenos/fisiologia , Células Cultivadas , Chlorocebus aethiops , Infecções por Coronavirus/imunologia , Aprovação de Drogas , Avaliação Pré-Clínica de Medicamentos , Humanos , Macrófagos/fisiologia , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Monócitos/fisiologia , Resultado do Tratamento , Células Vero
8.
PLoS One ; 13(3): e0194880, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29566079

RESUMO

Identifying effective antivirals for treating Ebola virus disease (EVD) and minimizing transmission of such disease is critical. A variety of cell-based assays have been developed for evaluating compounds for activity against Ebola virus. However, very few reports discuss the variable assay conditions that can affect the results obtained from these drug screens. Here, we describe variable conditions tested during the development of our cell-based drug screen assays designed to identify compounds with anti-Ebola virus activity using established cell lines and human primary cells. The effect of multiple assay readouts and variable assay conditions, including virus input, time of infection, and the cell passage number, were compared, and the impact on the effective concentration for 50% and/ or 90% inhibition (EC50, EC90) was evaluated using the FDA-approved compound, toremifene citrate. In these studies, we show that altering cell-based assay conditions can have an impact on apparent drug potency as measured by the EC50. These results further support the importance of developing standard operating procedures for generating reliable and reproducible in vitro data sets for potential antivirals.


Assuntos
Antivirais/uso terapêutico , Técnicas de Cultura de Células , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Doença pelo Vírus Ebola/tratamento farmacológico , Animais , Técnicas de Cultura de Células/métodos , Células Cultivadas , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos/normas , Ebolavirus/efeitos dos fármacos , Ebolavirus/fisiologia , Ensaios de Triagem em Larga Escala/métodos , Humanos , Resultado do Tratamento , Células Vero
9.
PLoS One ; 11(11): e0166318, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27902714

RESUMO

In the fall of 2014, an international news agency reported that patients suffering from Ebola virus disease (EVD) in Liberia were treated successfully with lamivudine, an antiviral drug used to treat human immunodeficiency virus-1 and hepatitis B virus infections. According to the report, 13 out of 15 patients treated with lamivudine survived and were declared free from Ebola virus disease. In this study, the anti-Ebola virus (EBOV) activity of lamivudine and another antiretroviral, zidovudine, were evaluated in a diverse set of cell lines against two variants of wild-type EBOV. Variable assay parameters were assessed to include different multiplicities of infection, lengths of inoculation times, and durations of dosing. At a multiplicity of infection of 1, lamivudine and zidovudine had no effect on EBOV propagation in Vero E6, Hep G2, or HeLa cells, or in primary human monocyte-derived macrophages. At a multiplicity of infection of 0.1, zidovudine demonstrated limited anti-EBOV activity in Huh 7 cells. Under certain conditions, lamivudine had low anti-EBOV activity at the maximum concentration tested (320 µM). However, lamivudine never achieved greater than 30% viral inhibition, and the activity was not consistently reproducible. Combination of lamivudine and zidovudine showed no synergistic antiviral activity. Independently, a set of in vitro experiments testing lamivudine and zidovudine for antiviral activity against an Ebola-enhanced green fluorescent protein reporter virus was performed at the Centers for Disease Control and Prevention. No antiviral activity was observed for either compound. A study evaluating the efficacy of lamivudine in a guinea pig model of EVD found no survival benefit. This lack of benefit was observed despite plasma lamivudine concentrations in guinea pig of about 4 µg/ml obtained in a separately conducted pharmacokinetics study. These studies found no evidence to support the therapeutic use of lamivudine for the treatment of EVD.


Assuntos
Fármacos Anti-HIV/farmacologia , Ebolavirus/efeitos dos fármacos , Doença pelo Vírus Ebola/tratamento farmacológico , Lamivudina/farmacologia , Zidovudina/farmacologia , Animais , Chlorocebus aethiops , Ebolavirus/isolamento & purificação , Cobaias , Células HeLa , Doença pelo Vírus Ebola/virologia , Humanos , Macrófagos , Projetos Piloto , Células Vero , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA