Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Res Sq ; 2023 Dec 22.
Article in English | MEDLINE | ID: mdl-38196595

ABSTRACT

Monoclonal antibodies (mAbs) against Ebola virus (EBOV) glycoprotein (GP1,2) are the standard of care for Ebola virus disease (EVD). Anti-GP1,2 mAbs targeting the stalk and membrane proximal external region (MPER) potently neutralize EBOV in vitro. However, their neutralization mechanism is poorly understood because they target a GP1,2 epitope that has evaded structural characterization. Moreover, their in vivo efficacy has only been evaluated in the mouse model of EVD. Using x-ray crystallography and cryo-electron tomography of 3A6 complexed with its stalk- GP1,2 MPER epitope we reveal a novel mechanism in which 3A6 elevates the stalk or stabilizes a conformation of GP1,2 that is lifted from the virion membrane. In domestic guinea pig and rhesus monkey EVD models, 3A6 provides therapeutic benefit at high viremia levels, advanced disease stages, and at the lowest dose yet demonstrated for any anti-EBOV mAb-based monotherapy. These findings can guide design of next-generation, highly potent anti-EBOV mAbs.

3.
Microorganisms ; 9(3)2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33801811

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-33468464

ABSTRACT

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.


Subject(s)
Antiviral Agents/therapeutic use , Arenaviridae Infections/drug therapy , Arenavirus/drug effects , Administration, Oral , Animals , Arenaviridae Infections/virology , Cell Line , Chlorocebus aethiops , Drug Synergism , Drug Therapy, Combination/methods , HEK293 Cells , Humans , Mice , Proof of Concept Study , Vero Cells
5.
Comp Med ; 70(1): 87-92, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31948513

ABSTRACT

Guinea pigs are a premier small animal model for infectious disease research, and chronic indwelling venous access ports may be used to facilitate various procedures. Here we report catheter-related lesions in 5 uninfected Dunkin-Hartley guinea pigs with chronic jugular vein catheters used for imaging studies. Three guinea pigs were found dead with no premonitory signs. At necropsy, there was severe bilateral pulmonary atelectasis due to 20 to 29 mL of pleural effusion resulting from catheter-related thrombosis and cranial vena cava syndrome. In addition, one of these 3 guinea pigs had a polymicrobial catheter infection with abscessation. A 4th clinically normal guinea pig was euthanized at the end of the study, having spontaneously lost its catheter 7 mo prior, and had 17 mL of pleural effusion. The 5th guinea pig was euthanized following pooling of contrast material around the distal catheter in the cranial vena cava on CT. By histology, affected animals had recent and remote thrombosis or fibrosis (or both) of the cranial vena cava and right atrial wall, with osseous and cartilaginous metaplasia. Cranial vena cava syndrome should be considered as a differential for dyspnea or death in chronically catheterized laboratory animals.


Subject(s)
Catheters, Indwelling/adverse effects , Guinea Pigs , Superior Vena Cava Syndrome/etiology , Animals , Catheterization, Central Venous/adverse effects , Catheters, Indwelling/veterinary , Female , Jugular Veins , Male , Superior Vena Cava Syndrome/veterinary
6.
Sci Rep ; 9(1): 20199, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882748

ABSTRACT

During the Ebola virus disease (EVD) epidemic in Western Africa (2013‒2016), antimalarial treatment was administered to EVD patients due to the high coexisting malaria burden in accordance with World Health Organization guidelines. In an Ebola treatment center in Liberia, EVD patients receiving the combination antimalarial artesunate-amodiaquine had a lower risk of death compared to those treated with artemether-lumefantrine. As artemether and artesunate are derivatives of artemisinin, the beneficial anti-Ebola virus (EBOV) effect observed could possibly be attributed to the change from lumefantrine to amodiaquine. Amodiaquine is a widely used antimalarial in the countries that experience outbreaks of EVD and, therefore, holds promise as an approved drug that could be repurposed for treating EBOV infections. We investigated the potential anti-EBOV effect of amodiaquine in a well-characterized nonhuman primate model of EVD. Using a similar 3-day antimalarial dosing strategy as for human patients, plasma concentrations of amodiaquine in healthy animals were similar to those found in humans. However, the treatment regimen did not result in a survival benefit or decrease of disease signs in EBOV-infected animals. While amodiaquine on its own failed to demonstrate efficacy, we cannot exclude potential therapeutic value of amodiaquine when used in combination with artesunate or another antiviral.


Subject(s)
Amodiaquine/therapeutic use , Antiviral Agents/therapeutic use , Artemisinins/therapeutic use , Hemorrhagic Fever, Ebola/drug therapy , Animals , Disease Models, Animal , Drug Combinations , Female , Macaca mulatta , Male
7.
PLoS Negl Trop Dis ; 13(7): e0007595, 2019 07.
Article in English | MEDLINE | ID: mdl-31356611

ABSTRACT

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.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Musa/chemistry , Plant Lectins/pharmacology , Animals , Antiviral Agents/chemical synthesis , Cell Line, Tumor , Ebolavirus/genetics , Ebolavirus/immunology , Escherichia coli , Female , Genetic Engineering , HEK293 Cells , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/prevention & control , Humans , Immunoglobulin Variable Region/genetics , Mice , Mice, Inbred C57BL , Plant Lectins/chemical synthesis , Virus Replication/drug effects
8.
Sci Rep ; 9(1): 7484, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31097731

ABSTRACT

Development of antiviral drug resistance is a continuous concern for viruses with high mutation rates such as influenza. The use of antiviral drugs targeting host proteins required for viral replication is less likely to result in the selection of resistant viruses than treating with direct-acting antivirals. The iminosugar UV-4B is a host-targeted glucomimetic that inhibits endoplasmic reticulum α-glucosidase I and II enzymes resulting in improper glycosylation and misfolding of viral glycoproteins. UV-4B has broad-spectrum antiviral activity against diverse viruses including dengue and influenza. To examine the ability of influenza virus to develop resistance against UV-4B, mouse-adapted influenza virus was passaged in mice in the presence or absence of UV-4B and virus isolated from lungs was used to infect the next cohort of mice, for five successive passages. Deep sequencing was performed to identify changes in the viral genome during passaging in the presence or absence of UV-4B. Relatively few minor variants were identified within each virus and the ratio of nonsynonymous to synonymous (dN/dS) substitutions of minor variants confirmed no apparent positive selection following sustained exposure to UV-4B. Three substitutions (one synonymous in PB2, one nonsynonymous in M and PA each) were specifically enriched (>3%) in UV-4B-treated groups at passage five. Recombinant viruses containing each individual or combinations of these nonsynonymous mutations remained sensitive to UV-4B treatment in mice. Overall, these data provide evidence that there is a high genetic barrier to the generation and selection of escape mutants following exposure to host-targeted iminosugar antivirals.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral/genetics , Glycoside Hydrolase Inhibitors/pharmacology , Influenza A virus/genetics , Orthomyxoviridae Infections/virology , Animals , Female , Genome, Viral , Influenza A virus/drug effects , Mice , Mice, Inbred BALB C , Mutation , Recombination, Genetic , Selection, Genetic
9.
J Infect Dis ; 219(11): 1818-1822, 2019 05 05.
Article in English | MEDLINE | ID: mdl-30517671

ABSTRACT

Lassa fever (LF) survivors develop various clinical manifestations including polyserositis, myalgia, epididymitis, and hearing loss weeks to months after recovery from acute infection. We demonstrate a systemic lymphoplasmacytic and histiocytic arteritis and periarteritis in guinea pigs more than 2 months after recovery from acute Lassa virus (LASV) infection. LASV was detected in the arterial tunica media smooth muscle cells by immunohistochemistry, in situ hybridization, and transmission electron microscopy. Our results suggest that the sequelae of LASV infection may be due to virus persistence resulting in systemic vascular damage. These findings shed light on the pathogenesis of LASV sequelae in convalescent human survivors.


Subject(s)
Lassa Fever/virology , Lassa virus/immunology , Animals , Convalescence , Disease Models, Animal , Disease Progression , Female , Guinea Pigs , Humans , Immunohistochemistry , Inflammation , Lassa Fever/pathology , Male
10.
Viruses ; 10(11)2018 11 20.
Article in English | MEDLINE | ID: mdl-30463334

ABSTRACT

Lassa virus (LASV), a mammarenavirus, infects an estimated 100,000⁻300,000 individuals yearly in western Africa and frequently causes lethal disease. Currently, no LASV-specific antivirals or vaccines are commercially available for prevention or treatment of Lassa fever, the disease caused by LASV. The development of medical countermeasure screening platforms is a crucial step to yield licensable products. Using reverse genetics, we generated a recombinant wild-type LASV (rLASV-WT) and a modified version thereof encoding a cleavable green fluorescent protein (GFP) as a reporter for rapid and quantitative detection of infection (rLASV-GFP). Both rLASV-WT and wild-type LASV exhibited similar growth kinetics in cultured cells, whereas growth of rLASV-GFP was slightly impaired. GFP reporter expression by rLASV-GFP remained stable over several serial passages in Vero cells. Using two well-characterized broad-spectrum antivirals known to inhibit LASV infection, favipiravir and ribavirin, we demonstrate that rLASV-GFP is a suitable screening tool for the identification of LASV infection inhibitors. Building on these findings, we established a rLASV-GFP-based high-throughput drug discovery screen and an rLASV-GFP-based antibody neutralization assay. Both platforms, now available as a standard tool at the IRF-Frederick (an international resource), will accelerate anti-LASV medical countermeasure discovery and reduce costs of antiviral screens in maximum containment laboratories.


Subject(s)
Drug Evaluation, Preclinical/methods , Genes, Reporter , Green Fluorescent Proteins/analysis , Lassa virus/growth & development , Luminescent Agents/analysis , Neutralization Tests/methods , Staining and Labeling/methods , Animals , Antibodies, Neutralizing/immunology , Antiviral Agents/pharmacology , Chlorocebus aethiops , Fluorometry/methods , Genomic Instability , Green Fluorescent Proteins/genetics , Lassa virus/drug effects , Lassa virus/genetics , Lassa virus/immunology , Reverse Genetics , Ribavirin/pharmacology , Vero Cells
11.
J Infect Dis ; 218(suppl_5): S592-S596, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30016444

ABSTRACT

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.


Subject(s)
Amiodarone/pharmacology , Ebolavirus/drug effects , Amiodarone/pharmacokinetics , Amiodarone/therapeutic use , Animals , Chlorocebus aethiops , Female , Guinea Pigs , Hemorrhagic Fever, Ebola/drug therapy , Male , Vero Cells
12.
J Infect Dis ; 218(suppl_5): S588-S591, 2018 11 22.
Article in English | MEDLINE | ID: mdl-29982632

ABSTRACT

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.


Subject(s)
Bepridil/therapeutic use , Calcium Channel Blockers/therapeutic use , Marburg Virus Disease/drug therapy , Animals , Chlorocebus aethiops , Disease Models, Animal , Female , Marburg Virus Disease/mortality , Mice , Mice, Inbred BALB C , Vero Cells
13.
J Infect Dis ; 218(suppl_5): S672-S678, 2018 11 22.
Article in English | MEDLINE | ID: mdl-29939303

ABSTRACT

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.


Subject(s)
Antiviral Agents/administration & dosage , Ebolavirus/drug effects , Animals , Antiviral Agents/therapeutic use , Chlorocebus aethiops , Drug Approval , Drug Synergism , Drug Therapy, Combination , Vero Cells , Virion/drug effects , Virus Internalization/drug effects
14.
PLoS One ; 13(3): e0194880, 2018.
Article in English | MEDLINE | ID: mdl-29566079

ABSTRACT

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.


Subject(s)
Antiviral Agents/therapeutic use , Cell Culture Techniques , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Hemorrhagic Fever, Ebola/drug therapy , Animals , Cell Culture Techniques/methods , Cells, Cultured , Chlorocebus aethiops , Drug Evaluation, Preclinical/standards , Ebolavirus/drug effects , Ebolavirus/physiology , High-Throughput Screening Assays/methods , Humans , Treatment Outcome , Vero Cells
15.
PLoS Negl Trop Dis ; 11(4): e0005540, 2017 04.
Article in English | MEDLINE | ID: mdl-28403145

ABSTRACT

Phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) is a lipid kinase involved in endosome maturation that emerged from a haploid genetic screen as being required for Ebola virus (EBOV) infection. Here we analyzed the effects of apilimod, a PIKfyve inhibitor that was reported to be well tolerated in humans in phase 2 clinical trials, for its effects on entry and infection of EBOV and Marburg virus (MARV). We first found that apilimod blocks infections by EBOV and MARV in Huh 7, Vero E6 and primary human macrophage cells, with notable potency in the macrophages (IC50, 10 nM). We next observed that similar doses of apilimod block EBOV-glycoprotein-virus like particle (VLP) entry and transcription-replication competent VLP infection, suggesting that the primary mode of action of apilimod is as an entry inhibitor, preventing release of the viral genome into the cytoplasm to initiate replication. After providing evidence that the anti-EBOV action of apilimod is via PIKfyve, we showed that it blocks trafficking of EBOV VLPs to endolysosomes containing Niemann-Pick C1 (NPC1), the intracellular receptor for EBOV. Concurrently apilimod caused VLPs to accumulate in early endosome antigen 1-positive endosomes. We did not detect any effects of apilimod on bulk endosome acidification, on the activity of cathepsins B and L, or on cholesterol export from endolysosomes. Hence by antagonizing PIKfyve, apilimod appears to block EBOV trafficking to its site of fusion and entry into the cytoplasm. Given the drug's observed anti-filoviral activity, relatively unexplored mechanism of entry inhibition, and reported tolerability in humans, we propose that apilimod be further explored as part of a therapeutic regimen to treat filoviral infections.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Marburgvirus/drug effects , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Triazines/pharmacology , Virus Internalization/drug effects , Animals , Biological Transport , Cell Line , Chlorocebus aethiops , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/drug therapy , Humans , Hydrazones , Lysosomes/metabolism , Macrophages/virology , Marburgvirus/physiology , Nocodazole/pharmacology , Pyrimidines , Toremifene/pharmacology , Vero Cells
16.
J Infect Dis ; 215(9): 1416-1420, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28368541

ABSTRACT

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.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Animals , Cell Line, Tumor , Chlorocebus aethiops , Drug Synergism , Humans , Vero Cells
17.
PLoS One ; 11(11): e0166318, 2016.
Article in English | MEDLINE | ID: mdl-27902714

ABSTRACT

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.


Subject(s)
Anti-HIV Agents/pharmacology , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/drug therapy , Lamivudine/pharmacology , Zidovudine/pharmacology , Animals , Chlorocebus aethiops , Ebolavirus/isolation & purification , Guinea Pigs , HeLa Cells , Hemorrhagic Fever, Ebola/virology , Humans , Macrophages , Pilot Projects , Vero Cells , Virus Replication/drug effects
18.
Sci Transl Med ; 7(290): 290ra89, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26041706

ABSTRACT

Currently, no approved therapeutics exist to treat or prevent infections induced by Ebola viruses, and recent events have demonstrated an urgent need for rapid discovery of new treatments. Repurposing approved drugs for emerging infections remains a critical resource for potential antiviral therapies. We tested ~2600 approved drugs and molecular probes in an in vitro infection assay using the type species, Zaire ebolavirus. Selective antiviral activity was found for 80 U.S. Food and Drug Administration-approved drugs spanning multiple mechanistic classes, including selective estrogen receptor modulators, antihistamines, calcium channel blockers, and antidepressants. Results using an in vivo murine Ebola virus infection model confirmed the protective ability of several drugs, such as bepridil and sertraline. Viral entry assays indicated that most of these antiviral drugs block a late stage of viral entry. By nature of their approved status, these drugs have the potential to be rapidly advanced to clinical settings and used as therapeutic countermeasures for Ebola virus infections.


Subject(s)
Antiviral Agents/therapeutic use , Drug Approval , Hemorrhagic Fever, Ebola/therapy , Molecular Probes , Animals , Bepridil/pharmacology , Ebolavirus/drug effects , Humans , Mice , Sertraline/pharmacology
19.
Sci Transl Med ; 5(190): 190ra79, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23785035

ABSTRACT

Ebola viruses remain a substantial threat to both civilian and military populations as bioweapons, during sporadic outbreaks, and from the possibility of accidental importation from endemic regions by infected individuals. Currently, no approved therapeutics exist to treat or prevent infection by Ebola viruses. Therefore, we performed an in vitro screen of Food and Drug Administration (FDA)- and ex-US-approved drugs and selected molecular probes to identify drugs with antiviral activity against the type species Zaire ebolavirus (EBOV). From this screen, we identified a set of selective estrogen receptor modulators (SERMs), including clomiphene and toremifene, which act as potent inhibitors of EBOV infection. Anti-EBOV activity was confirmed for both of these SERMs in an in vivo mouse infection model. This anti-EBOV activity occurred even in the absence of detectable estrogen receptor expression, and both SERMs inhibited virus entry after internalization, suggesting that clomiphene and toremifene are not working through classical pathways associated with the estrogen receptor. Instead, the response appeared to be an off-target effect where the compounds interfere with a step late in viral entry and likely affect the triggering of fusion. These data support the screening of readily available approved drugs to identify therapeutics for the Ebola viruses and other infectious diseases. The SERM compounds described in this report are an immediately actionable class of approved drugs that can be repurposed for treatment of filovirus infections.


Subject(s)
Drug Approval , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/drug therapy , Selective Estrogen Receptor Modulators/therapeutic use , United States Food and Drug Administration , Animals , Cathepsins/metabolism , Chlorocebus aethiops , Clomiphene/pharmacology , Clomiphene/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Ebolavirus/drug effects , Endosomes/drug effects , Endosomes/metabolism , Hemorrhagic Fever, Ebola/virology , Hep G2 Cells , Humans , Hydrogen-Ion Concentration/drug effects , Mice , Mice, Inbred C57BL , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Survival Analysis , Toremifene/pharmacology , Toremifene/therapeutic use , United States , Vero Cells , Virion/drug effects , Virus Internalization/drug effects
20.
J Neurosci ; 31(9): 3470-83, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21368059

ABSTRACT

The RE1-silencing transcription factor (REST) represses the expression of neuronal-specific genes in non-neuronal cells by recruiting histone deacetylases (HDACs) and other histone modifying and chromatin remodeling proteins to the DNA. REST regulation of the expression of neuronal genes is required for the orderly developmental transition from a neuroepithelial stem cell to a functional neuron. Here, we examined the expression and function of REST in neonatal rat oligodendrocyte precursor cells (OPCs). OPCs develop from the same neuroepithelial stem cells as neurons, can be reprogrammed to act as neural stem-like cells in vitro, and require HDAC-mediated gene repression to develop into mature oligodendrocytes. We show that OPCs express functional REST protein and that REST interacts with several neuronal-specific genes whose expression is repressed in OPCs. REST transcript and protein expression increased fourfold during the first 48 h of oligodendrocyte differentiation. During this differentiation, the expression of RE1 containing neuronal genes further decreased as the transcription of oligodendrocyte-specific genes was activated. Expression of a dominant-negative form of REST in OPCs prevented the cells from developing into mature MBP-positive oligodendrocytes. Rather, the cells began to develop a neuronal phenotype characterized by increased expression of neuronal proteins, transcription factors, and cell-type-specific marker antigens. REST overexpression promoted the development of O4-positive pre-oligodendrocytes from OPCs. Together, these results show that REST function is required for the differentiation of OPCs into oligodendrocytes. By regulating the expression of neuronal genes, REST may also regulate the phenotypic plasticity of OPCs.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/physiology , Oligodendroglia/cytology , Oligodendroglia/physiology , Repressor Proteins/physiology , Animals , Animals, Newborn , Cells, Cultured , Female , Male , Neuronal Plasticity/physiology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...