Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
mBio ; 12(6): e0334721, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34933447

RESUMO

The world was unprepared for coronavirus disease 2019 (COVID-19) and remains ill-equipped for future pandemics. While unprecedented strides have been made developing vaccines and treatments for COVID-19, there remains a need for highly effective and widely available regimens for ambulatory use for novel coronaviruses and other viral pathogens. We posit that a priority is to develop pan-family drug cocktails to enhance potency, limit toxicity, and avoid drug resistance. We urge cocktail development for all viruses with pandemic potential both in the short term (<1 to 2 years) and longer term with pairs of drugs in advanced clinical testing or repurposed agents approved for other indications. While significant efforts were launched against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), in vitro and in the clinic, many studies employed solo drugs and had disappointing results. Here, we review drug combination studies against SARS-CoV-2 and other viruses and introduce a model-driven approach to assess drug pairs with the highest likelihood of clinical efficacy. Where component agents lack sufficient potency, we advocate for synergistic combinations to achieve therapeutic levels. We also discuss issues that stymied therapeutic progress against COVID-19, including testing of agents with low likelihood of efficacy late in clinical disease and lack of focus on developing virologic surrogate endpoints. There is a need to expedite efficient clinical trials testing drug combinations that could be taken at home by recently infected individuals and exposed contacts as early as possible during the next pandemic, whether caused by a coronavirus or another viral pathogen. The approach herein represents a proactive plan for global viral pandemic preparedness.


Assuntos
Antivirais/farmacologia , Coronavirus/efeitos dos fármacos , Combinação de Medicamentos , Animais , Coronavirus/classificação , Coronavirus/patogenicidade , Infecções por Coronavirus/tratamento farmacológico , Humanos , Camundongos , Pandemias/prevenção & controle , SARS-CoV-2/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
3.
Microorganisms ; 9(3)2021 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-33801811

RESUMO

Outbreaks of Ebola ebolavirus (EBOV) have been associated with high morbidity and mortality. Milestones have been reached recently in the management of EBOV disease (EVD) with licensure of an EBOV vaccine and two monoclonal antibody therapies. However, neither vaccines nor therapies are available for other disease-causing filoviruses. In preparation for such outbreaks, and for more facile and cost-effective management of EVD, we seek a cocktail containing orally available and room temperature stable drugs with strong activity against multiple filoviruses. We previously showed that (bepridil + sertraline) and (sertraline + toremifene) synergistically suppress EBOV in cell cultures. Here, we describe steps towards testing these combinations in a mouse model of EVD. We identified a vehicle suitable for oral delivery of the component drugs and determined that, thus formulated the drugs are equally active against EBOV as preparations in DMSO, and they maintain activity upon storage in solution for up to seven days. Pharmacokinetic (PK) studies indicated that the drugs in the oral delivery vehicle are well tolerated in mice at the highest doses tested. Collectively the data support advancement of these combinations to tests for synergy in a mouse model of EVD. Moreover, mathematical modeling based on human oral PK projects that the combinations would be more active in humans than their component single drugs.

4.
Artigo em Inglês | MEDLINE | ID: mdl-33468464

RESUMO

Neglected diseases caused by arenaviruses such as Lassa virus (LASV) and filoviruses like Ebola virus (EBOV) primarily afflict resource-limited countries, where antiviral drug development is often minimal. Previous studies have shown that many approved drugs developed for other clinical indications inhibit EBOV and LASV and that combinations of these drugs provide synergistic suppression of EBOV, often by blocking discrete steps in virus entry. We hypothesize that repurposing of combinations of orally administered approved drugs provides effective suppression of arenaviruses. In this report, we demonstrate that arbidol, an approved influenza antiviral previously shown to inhibit EBOV, LASV, and many other viruses, inhibits murine leukemia virus (MLV) reporter viruses pseudotyped with the fusion glycoproteins (GPs) of other arenaviruses (Junin virus [JUNV], lymphocytic choriomeningitis virus [LCMV], and Pichinde virus [PICV]). Arbidol and other approved drugs, including aripiprazole, amodiaquine, sertraline, and niclosamide, also inhibit infection of cells by infectious PICV, and arbidol, sertraline, and niclosamide inhibit infectious LASV. Combining arbidol with aripiprazole or sertraline results in the synergistic suppression of LASV and JUNV GP-bearing pseudoviruses. This proof-of-concept study shows that arenavirus infection in vitro can be synergistically inhibited by combinations of approved drugs. This approach may lead to a proactive strategy with which to prepare for and control known and new arenavirus outbreaks.


Assuntos
Antivirais/uso terapêutico , Infecções por Arenaviridae/tratamento farmacológico , Arenavirus/efeitos dos fármacos , Administração Oral , Animais , Infecções por Arenaviridae/virologia , Linhagem Celular , Chlorocebus aethiops , Sinergismo Farmacológico , Quimioterapia Combinada/métodos , Células HEK293 , Humanos , Camundongos , Estudo de Prova de Conceito , Células Vero
5.
Science ; 370(6513): 241-247, 2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-32855215

RESUMO

Recent outbreaks of Ebola virus (EBOV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have exposed our limited therapeutic options for such diseases and our poor understanding of the cellular mechanisms that block viral infections. Using a transposon-mediated gene-activation screen in human cells, we identify that the major histocompatibility complex (MHC) class II transactivator (CIITA) has antiviral activity against EBOV. CIITA induces resistance by activating expression of the p41 isoform of invariant chain CD74, which inhibits viral entry by blocking cathepsin-mediated processing of the Ebola glycoprotein. We further show that CD74 p41 can block the endosomal entry pathway of coronaviruses, including SARS-CoV-2. These data therefore implicate CIITA and CD74 in host defense against a range of viruses, and they identify an additional function of these proteins beyond their canonical roles in antigen presentation.


Assuntos
Antígenos de Diferenciação de Linfócitos B/fisiologia , Betacoronavirus/fisiologia , Infecções por Coronavirus/imunologia , Ebolavirus/fisiologia , Doença pelo Vírus Ebola/imunologia , Antígenos de Histocompatibilidade Classe II/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Proteínas Nucleares/fisiologia , Pneumonia Viral/imunologia , Transativadores/fisiologia , Internalização do Vírus , Antígenos de Diferenciação de Linfócitos B/genética , COVID-19 , Linhagem Celular Tumoral , Infecções por Coronavirus/virologia , Elementos de DNA Transponíveis , Endossomos/virologia , Testes Genéticos , Doença pelo Vírus Ebola/virologia , Antígenos de Histocompatibilidade Classe II/genética , Interações Hospedeiro-Patógeno/genética , Humanos , Proteínas Nucleares/genética , Pandemias , Pneumonia Viral/virologia , SARS-CoV-2 , Transativadores/genética , Transcrição Gênica
6.
Adv Ther (Weinh) ; 3(7): 2000034, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32838027

RESUMO

In 2019/2020, the emergence of coronavirus disease 2019 (COVID-19) resulted in rapid increases in infection rates as well as patient mortality. Treatment options addressing COVID-19 included drug repurposing, investigational therapies such as remdesivir, and vaccine development. Combination therapy based on drug repurposing is among the most widely pursued of these efforts. Multi-drug regimens are traditionally designed by selecting drugs based on their mechanism of action. This is followed by dose-finding to achieve drug synergy. This approach is widely-used for drug development and repurposing. Realizing synergistic combinations, however, is a substantially different outcome compared to globally optimizing combination therapy, which realizes the best possible treatment outcome by a set of candidate therapies and doses toward a disease indication. To address this challenge, the results of Project IDentif.AI (Identifying Infectious Disease Combination Therapy with Artificial Intelligence) are reported. An AI-based platform is used to interrogate a massive 12 drug/dose parameter space, rapidly identifying actionable combination therapies that optimally inhibit A549 lung cell infection by vesicular stomatitis virus within three days of project start. Importantly, a sevenfold difference in efficacy is observed between the top-ranked combination being optimally and sub-optimally dosed, demonstrating the critical importance of ideal drug and dose identification. This platform is disease indication and disease mechanism-agnostic, and potentially applicable to the systematic N-of-1 and population-wide design of highly efficacious and tolerable clinical regimens. This work also discusses key factors ranging from healthcare economics to global health policy that may serve to drive the broader deployment of this platform to address COVID-19 and future pandemics.

7.
PLoS Negl Trop Dis ; 13(7): e0007595, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31356611

RESUMO

Ebolaviruses cause an often rapidly fatal syndrome known as Ebola virus disease (EVD), with average case fatality rates of ~50%. There is no licensed vaccine or treatment for EVD, underscoring the urgent need to develop new anti-ebolavirus agents, especially in the face of an ongoing outbreak in the Democratic Republic of the Congo and the largest ever outbreak in Western Africa in 2013-2016. Lectins have been investigated as potential antiviral agents as they bind glycans present on viral surface glycoproteins, but clinical use of them has been slowed by concerns regarding their mitogenicity, i.e. ability to cause immune cell proliferation. We previously engineered a banana lectin (BanLec), a carbohydrate-binding protein, such that it retained antiviral activity but lost mitogenicity by mutating a single amino acid, yielding H84T BanLec (H84T). H84T shows activity against viruses containing high-mannose N-glycans, including influenza A and B, HIV-1 and -2, and hepatitis C virus. Since ebolavirus surface glycoproteins also contain many high-mannose N-glycans, we assessed whether H84T could inhibit ebolavirus replication. H84T inhibited Ebola virus (EBOV) replication in cell cultures. In cells, H84T inhibited both virus-like particle (VLP) entry and transcription/replication of the EBOV mini-genome at high micromolar concentrations, while inhibiting infection by transcription- and replication-competent VLPs, which measures the full viral life cycle, in the low micromolar range. H84T did not inhibit assembly, budding, or release of VLPs. These findings suggest that H84T may exert its anti-ebolavirus effect(s) by blocking both entry and transcription/replication. In a mouse model, H84T partially (maximally, ~50-80%) protected mice from an otherwise lethal mouse-adapted EBOV infection. Interestingly, a single dose of H84T pre-exposure to EBOV protected ~80% of mice. Thus, H84T shows promise as a new anti-ebolavirus agent with potential to be used in combination with vaccination or other agents in a prophylactic or therapeutic regimen.


Assuntos
Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Musa/química , Lectinas de Plantas/farmacologia , Animais , Antivirais/síntese química , Linhagem Celular Tumoral , Ebolavirus/genética , Ebolavirus/imunologia , Escherichia coli , Feminino , Engenharia Genética , Células HEK293 , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/prevenção & controle , Humanos , Região Variável de Imunoglobulina/genética , Camundongos , Camundongos Endogâmicos C57BL , Lectinas de Plantas/síntese química , Replicação Viral/efeitos dos fármacos
8.
Viruses ; 11(2)2019 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-30781518

RESUMO

Following the largest Ebola virus disease outbreak from 2013 to 2016, viral RNA has been detected in survivors from semen and breast milk long after disease recovery. However, as there have been few cases of sexual transmission, it is unclear whether every RNA positive fluid sample contains infectious virus. Virus isolation, typically using cell culture or animal models, can serve as a tool to determine the infectivity of patient samples. However, the sensitivity of these methods has not been assessed for the Ebola virus isolate, Makona. Described here is an efficiency comparison of Ebola virus Makona isolation using Vero E6, Huh-7, monocyte-derived macrophage cells, and suckling laboratory mice. Isolation sensitivity was similar in all methods tested. Laboratory mice and Huh-7 cells were less affected by toxicity from breast milk than Vero E6 and MDM cells. However, the advantages associated with isolation in Huh-7 cells over laboratory mice, including cost effectiveness, sample volume preservation, and a reduction in animal use, make Huh-7 cells the preferred substrate tested for Ebola virus Makona isolation.


Assuntos
Ebolavirus/isolamento & purificação , Virologia/métodos , Animais , Animais Lactentes , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Doença pelo Vírus Ebola/virologia , Humanos , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Leite Humano/virologia , Sêmen/virologia , Sensibilidade e Especificidade , Células Vero
9.
Trop Med Infect Dis ; 3(2)2018 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-30274451

RESUMO

The World Health Organization (WHO) estimates that zoonotic diseases transmitted from animals to humans account for 75 percent of new and emerging infectious diseases. Globally, high-consequence pathogens that impact livestock and have the potential for human transmission create research paradoxes and operational challenges for the high-containment laboratories that conduct work with them. These specialized facilities are required for conducting all phases of research on high-consequence pathogens (basic, applied, and translational) with an emphasis on both the generation of fundamental knowledge and product development. To achieve this research mission, a highly-trained workforce is required and flexible operational methods are needed. In addition, working with certain pathogens requires compliance with regulations such as the Centers for Disease Control (CDC) and the U.S. Department of Agriculture (USDA) Select Agent regulations, which adds to the operational burden. The vast experience from the existing studies at Plum Island Animal Disease Center, other U.S. laboratories, and those in Europe and Australia with biosafety level 4 (BSL-4) facilities designed for large animals, clearly demonstrates the valuable contribution this capability brings to the efforts to detect, prepare, prevent and respond to livestock and potential zoonotic threats. To raise awareness of these challenges, which include biosafety and biosecurity issues, we held a workshop at the 2018 American Society for Microbiology (ASM) Biothreats conference to further discuss the topic with invited experts and audience participants. The workshop covered the subjects of research funding and metrics, economic sustainment of drug and vaccine development pipelines, workforce turnover, and the challenges of maintaining operational readiness of high containment laboratories.

10.
J Infect Dis ; 218(suppl_5): S636-S648, 2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-30010950

RESUMO

Transchromosomic bovines (Tc-bovines) adaptively produce fully human polyclonal immunoglobulin (Ig)G antibodies after exposure to immunogenic antigen(s). The National Interagency Confederation for Biological Research and collaborators rapidly produced and then evaluated anti-Ebola virus IgG immunoglobulins (collectively termed SAB-139) purified from Tc-bovine plasma after sequential hyperimmunization with an Ebola virus Makona isolate glycoprotein nanoparticle vaccine. SAB-139 was characterized by several in vitro production, research, and clinical level assays using wild-type Makona-C05 or recombinant virus/antigens from different Ebola virus variants. SAB-139 potently activates natural killer cells, monocytes, and peripheral blood mononuclear cells and has high-binding avidity demonstrated by surface plasmon resonance. SAB-139 has similar concentrations of galactose-α-1,3-galactose carbohydrates compared with human-derived intravenous Ig, and the IgG1 subclass antibody is predominant. All rhesus macaques infected with Ebola virus/H.sapiens-tc/GIN/2014/Makona-C05 and treated with sufficient SAB-139 at 1 day (n = 6) or 3 days (n = 6) postinfection survived versus 0% of controls. This study demonstrates that Tc-bovines can produce pathogen-specific human Ig to prevent and/or treat patients when an emerging infectious disease either threatens to or becomes an epidemic.


Assuntos
Anticorpos Antivirais/uso terapêutico , Ebolavirus/imunologia , Doença pelo Vírus Ebola/tratamento farmacológico , Imunoglobulina G/uso terapêutico , Animais , Bovinos , Chlorocebus aethiops , Feminino , Humanos , Macaca mulatta , Masculino , Células Vero
11.
J Infect Dis ; 218(suppl_5): S588-S591, 2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-29982632

RESUMO

No therapeutics are approved for the treatment of filovirus infections. Bepridil, a calcium channel blocker developed for treating angina, was identified as a potent inhibitor of filoviruses in vitro, including Ebola and Marburg viruses, and Ebola virus in vivo. We evaluated the efficacy of bepridil in a lethal mouse model of Marburg virus disease. A dose of 12 mg/kg bepridil once or twice daily resulted in 80% or 90% survival, respectively. These data confirm bepridil's broad-spectrum anti-filovirus activity warranting further investigation of bepridil, or improved compounds with a similar mechanism, as a pan-filovirus therapeutic agent.


Assuntos
Bepridil/uso terapêutico , Bloqueadores dos Canais de Cálcio/uso terapêutico , Doença do Vírus de Marburg/tratamento farmacológico , Animais , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Doença do Vírus de Marburg/mortalidade , Camundongos , Camundongos Endogâmicos BALB C , Células Vero
12.
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
13.
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
15.
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
16.
J Virol Methods ; 255: 84-90, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29481881

RESUMO

As part of the scientific community's development of medical countermeasures against Ebola virus disease, optimization of standardized assays for product evaluation is paramount. The recent outbreak heightened awareness to the scarcity of available assays and limited information on performance and reproducibility. To evaluate the immunogenicity of vaccines entering Phase I-III trials and to identify survivors, two enzyme-linked immunosorbent assays, the Filovirus Animal Non-Clinical Group assay and the Alpha Diagnostics International assay, were evaluated for detection of immunoglobulin G against Ebola virus glycoprotein. We found that the Filovirus Animal Nonclinical Group assay produced a wider range of relative antibody concentrations, higher assay precision, larger relative accuracy range, and lower regional background. Additionally, to sufficiently power a vaccine trial, use of the Filovirus Animal Nonclinical Group assay would require one third the number of participants than the Alpha Diagnostics International assay. This reduction in needed study participants will require less money, fewer man hours, and much less time to evaluate vaccine immunogenicity.


Assuntos
Ebolavirus , Doença pelo Vírus Ebola/diagnóstico , Imunoensaio , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Ebolavirus/imunologia , Ensaio de Imunoadsorção Enzimática , Filoviridae/imunologia , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/virologia , Humanos , Imunoensaio/métodos , Testes Sorológicos , Vacinas Virais/imunologia
17.
Drugs ; 77(18): 1935-1966, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29143192

RESUMO

No specific antivirals are currently available for two emerging infectious diseases, Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS). A literature search was performed covering pathogenesis, clinical features and therapeutics, clinically developed drugs for repurposing and novel drug targets. This review presents current knowledge on the epidemiology, pathogenesis and clinical features of the SARS and MERS coronaviruses. The rationale for and outcomes with treatments used for SARS and MERS is discussed. The main focus of the review is on drug development and the potential that drugs approved for other indications provide for repurposing. The drugs we discuss belong to a wide range of different drug classes, such as cancer therapeutics, antipsychotics, and antimalarials. In addition to their activity against MERS and SARS coronaviruses, many of these approved drugs have broad-spectrum potential and have already been in clinical use for treating other viral infections. A wealth of knowledge is available for these drugs. However, the information in this review is not meant to guide clinical decisions, and any therapeutic described here should only be used in context of a clinical trial. Potential targets for novel antivirals and antibodies are discussed as well as lessons learned from treatment development for other RNA viruses. The article concludes with a discussion of the gaps in our knowledge and areas for future research on emerging coronaviruses.


Assuntos
Antivirais/uso terapêutico , Infecções por Coronavirus/tratamento farmacológico , Síndrome Respiratória Aguda Grave/tratamento farmacológico , Antivirais/química , Infecções por Coronavirus/virologia , Aprovação de Drogas , Humanos , Coronavírus da Síndrome Respiratória do Oriente Médio , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave , Síndrome Respiratória Aguda Grave/virologia
18.
Sci Rep ; 7(1): 5886, 2017 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-28725019

RESUMO

The recent epidemic of Ebola virus disease in West Africa resulted in an unprecedented number of cases and deaths. Due to the scope of the outbreak combined with the lack of available approved treatment options, there was strong motivation to investigate any potential drug which had existing data reporting anti-Ebola activity. Drugs with demonstrated antiviral activity in the nonhuman primate models already approved for another indication or for which there was existing safety data were considered to be priorities for evaluation by the World Health Organization. Sertraline hydrochloride was reported to have anti-Ebola activity in vitro alone and in combination with other approved drugs. Although the efficacy was less than 100% in the murine model, the established safety profile of this product, the potential benefit alone and in combination, as well as the lack of other available options prioritized this compound for testing in the Ebola virus intramuscular rhesus macaque challenge model. Using a blinded dosing strategy, we demonstrated that high dose sertraline monotherapy provided no benefit for the prevention of Ebola virus disease in rhesus macaques with regards to reduction of viral load, morbidity, or survival highlighting the challenges of translating results between in vitro and in vivo models.


Assuntos
Ebolavirus/fisiologia , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/prevenção & controle , Sertralina/uso terapêutico , Animais , Relação Dose-Resposta a Droga , Ebolavirus/efeitos dos fármacos , Doença pelo Vírus Ebola/sangue , Doença pelo Vírus Ebola/patologia , Macaca mulatta , Masculino , Neutrófilos/patologia , Contagem de Plaquetas , Sertralina/administração & dosagem , Análise de Sobrevida , Carga Viral
19.
Curr Top Microbiol Immunol ; 411: 171-193, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28643203

RESUMO

Ebola virus disease (EVD) in humans is associated with four ebolaviruses: Ebola virus (EBOV), Sudan virus (SUDV), Bundibugyo virus (BDBV), and Taï Forest virus. To date, no documented cases of human disease have been associated with Reston virus. Here, we describe the nonhuman primate (NHP) models that currently serve as gold standards for testing ebolavirus vaccines and therapeutic agents and elucidating underlying mechanisms of pathogenesis. Although multiple models have been explored over the past 50 years, the predominance of published work has been performed in macaque models. This chapter will focus on the most commonly used models.


Assuntos
Modelos Animais de Doenças , Ebolavirus , Doença pelo Vírus Ebola , Macaca/virologia , Animais , Ebolavirus/classificação , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/terapia , Doença pelo Vírus Ebola/virologia , Humanos
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA