Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Vaccines (Basel) ; 11(6)2023 Jun 19.
Article in English | MEDLINE | ID: mdl-37376509

ABSTRACT

Outbreaks of viral hemorrhagic fever caused by filoviruses have become more prevalent in recent years, with outbreaks of Ebola virus (EBOV), Sudan virus (SUDV), and Marburg virus (MARV) all occurring in 2022 and 2023. While licensed vaccines are now available for EBOV, vaccine candidates for SUDV and MARV are all in preclinical or early clinical development phases. During the recent outbreak of SUDV virus disease, the Biomedical Advanced Research and Development Authority (BARDA), as part of the Administration for Strategic Preparedness and Response within the U.S. Department of Health and Human Services, implemented key actions with our existing partners to advance preparedness and enable rapid response to the outbreak, while also aligning with global partners involved in the implementation of clinical trials in an outbreak setting. Beyond pre-existing plans prior to the outbreak, BARDA worked with product sponsors to expedite manufacturing of vaccine doses that could be utilized in clinical trials. While the SUDV outbreak has since ended, a new outbreak of MARV disease has emerged. It remains critical that we continue to advance a portfolio of vaccines against SUDV and MARV while also expediting manufacturing activities ahead of, or in parallel if needed, outbreaks.

2.
Viruses ; 14(10)2022 10 18.
Article in English | MEDLINE | ID: mdl-36298846

ABSTRACT

The Biomedical Advanced Research and Development Authority, part of the Administration for Strategic Preparedness and Response within the U.S. Department of Health and Human Services, recognizes that the evaluation of medical countermeasures under the Animal Rule requires well-characterized and reproducible animal models that are likely to be predictive of clinical benefit. Marburg virus (MARV), one of two members of the genus Marburgvirus, is characterized by a hemorrhagic fever and a high case fatality rate for which there are no licensed vaccines or therapeutics available. This natural history study consisted of twelve cynomolgus macaques challenged with 1000 PFU of MARV Angola and observed for body weight, temperature, viremia, hematology, clinical chemistry, and coagulation at multiple time points. All animals succumbed to disease within 8 days and exhibited signs consistent with those observed in human cases, including viremia, fever, systemic inflammation, coagulopathy, and lymphocytolysis, among others. Additionally, this study determined the time from exposure to onset of disease manifestations and the time course, frequency, and magnitude of the manifestations. This study will be instrumental in the design and development of medical countermeasures to Marburg virus disease.


Subject(s)
Marburg Virus Disease , Marburgvirus , Medical Countermeasures , Humans , Animals , Marburgvirus/physiology , Viremia , Macaca fascicularis
3.
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
4.
Lancet Infect Dis ; 18(9): 1015-1024, 2018 09.
Article in English | MEDLINE | ID: mdl-30049622

ABSTRACT

BACKGROUND: Outbreak response efforts for the 2014-15 Ebola virus disease epidemic in west Africa brought widespread transmission to an end. However, subsequent clusters of infection have occurred in the region. An Ebola virus disease cluster in Liberia in November, 2015, that was identified after a 15-year-old boy tested positive for Ebola virus infection in Monrovia, raised the possibility of transmission from a persistently infected individual. METHODS: Case investigations were done to ascertain previous contact with cases of Ebola virus disease or infection with Ebola virus. Molecular investigations on blood samples explored a potential linkage between Ebola virus isolated from cases in this November, 2015, cluster and epidemiologically linked cases from the 2014-15 west African outbreak, according to the national case database. FINDINGS: The cluster investigated was the family of the index case (mother, father, three siblings). Ebola virus genomes assembled from two cases in the November, 2015, cluster, and an epidemiologically linked Ebola virus disease case in July, 2014, were phylogenetically related within the LB5 sublineage that circulated in Liberia starting around August, 2014. Partial genomes from two additional individuals, one from each cluster, were also consistent with placement in the LB5 sublineage. Sequencing data indicate infection with a lineage of the virus from a former transmission chain in the country. Based on serology and epidemiological and genomic data, the most plausible scenario is that a female case in the November, 2015, cluster survived Ebola virus disease in 2014, had viral persistence or recurrent disease, and transmitted the virus to three family members a year later. INTERPRETATION: Investigation of the source of infection for the November, 2015, cluster provides evidence of Ebola virus persistence and highlights the risk for outbreaks after interruption of active transmission. These findings underscore the need for focused prevention efforts among survivors and sustained capacity to rapidly detect and respond to new Ebola virus disease cases to prevent recurrence of a widespread outbreak. FUNDING: US Centers for Disease Control and Prevention, Defense Threat Reduction Agency, and WHO.


Subject(s)
Disease Outbreaks/prevention & control , Disease Outbreaks/statistics & numerical data , Epidemics/prevention & control , Epidemics/statistics & numerical data , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/transmission , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Infant , Liberia/epidemiology , Male , Middle Aged
5.
J Virol Methods ; 255: 84-90, 2018 05.
Article in English | MEDLINE | ID: mdl-29481881

ABSTRACT

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.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola/diagnosis , Immunoassay , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Ebolavirus/immunology , Enzyme-Linked Immunosorbent Assay , Filoviridae/immunology , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/virology , Humans , Immunoassay/methods , Serologic Tests , Viral Vaccines/immunology
6.
N Engl J Med ; 377(15): 1438-1447, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29020589

ABSTRACT

BACKGROUND: The safety and efficacy of vaccines to prevent Ebola virus disease (EVD) were unknown when the incidence of EVD was peaking in Liberia. METHODS: We initiated a randomized, placebo-controlled, phase 3 trial of the chimpanzee adenovirus 3 vaccine (ChAd3-EBO-Z) and the recombinant vesicular stomatitis virus vaccine (rVSV∆G-ZEBOV-GP) in Liberia. A phase 2 subtrial was embedded to evaluate safety and immunogenicity. Because the incidence of EVD declined in Liberia, the phase 2 component was expanded and the phase 3 component was eliminated. RESULTS: A total of 1500 adults underwent randomization and were followed for 12 months. The median age of the participants was 30 years; 36.6% of the participants were women. During the week after the administration of vaccine or placebo, adverse events occurred significantly more often with the active vaccines than with placebo; these events included injection-site reactions (in 28.5% of the patients in the ChAd3-EBO-Z group and 30.9% of those in the rVSV∆G-ZEBOV-GP group, as compared with 6.8% of those in the placebo group), headache (in 25.1% and 31.9%, vs. 16.9%), muscle pain (in 22.3% and 26.9%, vs. 13.3%), feverishness (in 23.9% and 30.5%, vs. 9.0%), and fatigue (in 14.0% and 15.4%, vs. 8.8%) (P<0.001 for all comparisons); these differences were not seen at 1 month. Serious adverse events within 12 months after injection were seen in 40 participants (8.0%) in the ChAd3-EBO-Z group, in 47 (9.4%) in the rVSV∆G-ZEBOV-GP group, and in 59 (11.8%) in the placebo group. By 1 month, an antibody response developed in 70.8% of the participants in the ChAd3-EBO-Z group and in 83.7% of those in the rVSV∆G-ZEBOV-GP group, as compared with 2.8% of those in the placebo group (P<0.001 for both comparisons). At 12 months, antibody responses in participants in the ChAd3-EBO-Z group (63.5%) and in those in the rVSV∆G-ZEBOV-GP group (79.5%) remained significantly greater than in those in the placebo group (6.8%, P<0.001 for both comparisons). CONCLUSIONS: A randomized, placebo-controlled phase 2 trial of two vaccines that was rapidly initiated and completed in Liberia showed the capability of conducting rigorous research during an outbreak. By 1 month after vaccination, the vaccines had elicited immune responses that were largely maintained through 12 months. (Funded by the National Institutes of Allergy and Infectious Diseases and the Liberian Ministry of Health; PREVAIL I ClinicalTrials.gov number, NCT02344407 .).


Subject(s)
Ebola Vaccines/adverse effects , Ebola Vaccines/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Adenoviridae , Adult , Animals , Disease Outbreaks , Double-Blind Method , Female , Fever/etiology , HIV Seropositivity/complications , Headache/etiology , Hemorrhagic Fever, Ebola/complications , Hemorrhagic Fever, Ebola/immunology , Humans , Injections, Intramuscular/adverse effects , Liberia , Male , Myalgia/etiology , Pan troglodytes , RNA, Viral/blood , Reverse Transcriptase Polymerase Chain Reaction , Vesiculovirus
8.
J Infect Dis ; 215(4): 547-553, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28003349

ABSTRACT

Recent studies have suggested that Ebola virus (EBOV) ribonucleic acid (RNA) potentially present in the semen of a large number of survivors of Ebola virus disease (EVD) in Western Africa may contribute to sexual transmission of EVD and generate new clusters of cases in regions previously declared EVD-free. These findings drive the immediate need for a reliable, rapid, user-friendly assay for detection of EBOV RNA in semen that is deployable to multiple sites across Western Africa. In this study, we optimized the Xpert EBOV assay for semen samples by adding dithiothreitol. Compared to the assays currently in use in Liberia (including Ebola Zaire Target 1, major groove binder real-time-polymerase chain reaction assays, and original Xpert EBOV assay), the modified Xpert EBOV assay demonstrated greater sensitivity than the comparator assays. Thus, the modified Xpert EBOV assay is optimal for large-scale monitoring of EBOV RNA persistence in male survivors.


Subject(s)
Ebolavirus/isolation & purification , Hemorrhagic Fever, Ebola/diagnosis , RNA, Viral/isolation & purification , Real-Time Polymerase Chain Reaction/methods , Semen/virology , Democratic Republic of the Congo , Hemorrhagic Fever, Ebola/blood , Humans , Liberia , Limit of Detection , Male , Pilot Projects , RNA, Viral/blood , Sensitivity and Specificity , Survivors
9.
Viruses ; 7(5): 2404-27, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25984714

ABSTRACT

Iminosugars are capable of targeting the life cycles of multiple viruses by blocking host endoplasmic reticulum α-glucosidase enzymes that are required for competent replication of a variety of enveloped, glycosylated viruses. Iminosugars as a class are approved for use in humans with diseases such as diabetes and Gaucher's disease, providing evidence for safety of this class of compounds. The in vitro antiviral activity of iminosugars has been described in several publications with a subset of these demonstrating in vivo activity against flaviviruses, herpesviruses, retroviruses and filoviruses. Although there is compelling non-clinical in vivo evidence of antiviral efficacy, the efficacy of iminosugars as antivirals has yet to be demonstrated in humans. In the current study, we report a novel iminosugar, UV-12, which has efficacy against dengue and influenza in mouse models. UV-12 exhibits drug-like properties including oral bioavailability and good safety profile in mice and guinea pigs. UV-12 is an example of an iminosugar with activity against multiple virus families that should be investigated in further safety and efficacy studies and demonstrates potential value of this drug class as antiviral therapeutics.


Subject(s)
Antiviral Agents/therapeutic use , Dengue/drug therapy , Imino Sugars/therapeutic use , Orthomyxoviridae Infections/drug therapy , Animals , Antiviral Agents/pharmacology , Dengue Virus/drug effects , Disease Models, Animal , Female , Guinea Pigs , Imino Sugars/pharmacology , Male , Mice , Microbial Sensitivity Tests , Orthomyxoviridae/drug effects , Treatment Outcome
10.
PLoS One ; 10(3): e0121662, 2015.
Article in English | MEDLINE | ID: mdl-25786028

ABSTRACT

Our lead iminosugar analog called UV-4 or N-(9-methoxynonyl)-1-deoxynojirimycin inhibits activity of endoplasmic reticulum (ER) α-glucosidases I and II and is a potent, host-targeted antiviral candidate. The mechanism of action for the antiviral activity of iminosugars is proposed to be inhibition of ER α-glucosidases leading to misfolding of critical viral glycoproteins. These misfolded glycoproteins would then be incorporated into defective virus particles or targeted for degradation resulting in a reduction of infectious progeny virions. UV-4, and its hydrochloride salt known as UV-4B, is highly potent against dengue virus in vitro and promotes complete survival in a lethal dengue virus mouse model. In the current studies, UV-4 was shown to be highly efficacious via oral gavage against both oseltamivir-sensitive and -resistant influenza A (H1N1) infections in mice even if treatment was initiated as late as 48-72 hours after infection. The minimal effective dose was found to be 80-100 mg/kg when administered orally thrice daily. UV-4 treatment did not affect the development of protective antibody responses after either influenza infection or vaccination. Therefore, UV-4 is a promising candidate for further development as a therapeutic intervention against influenza.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Antiviral Agents/pharmacology , Drug Resistance, Viral/drug effects , Influenza A Virus, H1N1 Subtype/drug effects , Oseltamivir/pharmacology , 1-Deoxynojirimycin/adverse effects , 1-Deoxynojirimycin/pharmacology , Animals , Antibodies, Viral/biosynthesis , Antiviral Agents/adverse effects , Female , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/physiology , Lung/drug effects , Male , Mice , Mice, Inbred BALB C , Safety , Time Factors , Viral Load/drug effects
11.
Antimicrob Agents Chemother ; 59(1): 85-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25313218

ABSTRACT

Dengue virus (DENV), a member of the Flaviviridae family, is a mosquito-borne pathogen and the cause of dengue fever. The increasing prevalence of DENV worldwide heightens the need for an effective vaccine and specific antivirals. Due to the dependence of DENV upon the lipid biosynthetic machinery of the host cell, lipid signaling and metabolism present unique opportunities for inhibiting viral replication. We screened a library of bioactive lipids and modulators of lipid metabolism and identified 4-hydroxyphenyl retinamide (4-HPR) (fenretinide) as an inhibitor of DENV in cell culture. 4-HPR inhibits the steady-state accumulation of viral genomic RNA and reduces viremia when orally administered in a murine model of DENV infection. The molecular target responsible for this antiviral activity is distinct from other known inhibitors of DENV but appears to affect other members of the Flaviviridae, including the West Nile, Modoc, and hepatitis C viruses. Although long-chain ceramides have been implicated in DENV replication, we demonstrate that DENV is insensitive to the perturbation of long-chain ceramides in mammalian cell culture and that the effect of 4-HPR on dihydroceramide homeostasis is separable from its antiviral activity. Likewise, the induction of reactive oxygen species by 4-HPR is not required for the inhibition of DENV. The inhibition of DENV in vivo by 4-HPR, combined with its well-established safety and tolerability in humans, suggests that it may be repurposed as a pan-Flaviviridae antiviral agent. This work also illustrates the utility of bioactive lipid screens for identifying critical interactions of DENV and other viral pathogens with host lipid biosynthesis, metabolism, and signal transduction.


Subject(s)
Dengue Virus/growth & development , Dengue/drug therapy , Fenretinide/therapeutic use , Viremia/drug therapy , Virus Replication/drug effects , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Female , HEK293 Cells , Hepacivirus/growth & development , Humans , Mice , Mice, Transgenic , Reactive Oxygen Species/metabolism , Vero Cells , West Nile virus/growth & development
12.
Toxicon ; 92: 36-41, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25260254

ABSTRACT

Due to the fast-acting nature of ricin, staphylococcal enterotoxin B (SEB), and Clostridium perfringens epsilon toxin (ETX), it is necessary that therapeutic interventions following a bioterrorism incident by one of these toxins occur as soon as possible after intoxication. Moreover, because the clinical manifestations of intoxication by these agents are likely to be indistinguishable from each other, especially following aerosol exposure, we have developed a cocktail of chimeric monoclonal antibodies that is capable of neutralizing all three toxins. The efficacy of this cocktail was demonstrated in mouse models of lethal dose toxin challenge.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bacterial Toxins/toxicity , Bioterrorism/prevention & control , Enterotoxins/toxicity , Poisoning/prevention & control , Recombinant Fusion Proteins/pharmacology , Ricin/toxicity , Animals , Female , Mice , Mice, Inbred BALB C , Neutralization Tests
13.
Antiviral Res ; 98(1): 35-43, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23376501

ABSTRACT

The aim of the present study was to evaluate the ability of the iminosugar drug UV-4 to provide in vivo protection from lethal dengue virus (DENV) challenge. This study utilized a well-described model of dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS)-like lethal disease in AG129 mice lacking the type I and II interferon receptors. Herein, we present UV-4 as a potent iminosugar for controlling DENV infection and disease in this mouse model. Specifically, administration of UV-4 reduced mortality, as well as viremia and viral RNA in key tissues, and cytokine storm. In addition, UV-4 treatment can be delayed, and it does not alter the anti-DENV antibody response. These results have set the foundation for development of UV-4 as a DENV-specific antiviral in phase I human clinical trials.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Dengue/drug therapy , Dengue/virology , Imino Sugars/pharmacology , Animals , Antiviral Agents/chemistry , Cytokines , Dengue/immunology , Dengue Virus/physiology , Humans , Imino Sugars/chemistry , Mice , Mice, Inbred Strains , Structure-Activity Relationship
14.
Antimicrob Agents Chemother ; 57(4): 1902-12, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23403421

ABSTRACT

Dengue virus (DENV) is the predominant mosquito-borne viral pathogen that infects humans with an estimated 50 to 100 million infections per year worldwide. Over the past 50 years, the incidence of dengue disease has increased dramatically and the virus is now endemic in more than 100 countries. Moreover, multiple serotypes of DENV are now found in the same geographic region, increasing the likelihood of more severe forms of disease. Despite extensive research, there are still no approved vaccines or therapeutics commercially available to treat DENV infection. Here we report the results of a high-throughput screen of a chemical compound library using a whole-virus assay that identified a novel small-molecule inhibitor of DENV, ST-610, that potently and selectively inhibits all four serotypes of DENV replication in vitro. Sequence analysis of drug-resistant virus isolates has identified a single point mutation, A263T, in the NS3 helicase domain that confers resistance to this compound. ST-610 inhibits DENV NS3 helicase RNA unwinding activity in a molecular-beacon-based helicase assay but does not inhibit nucleoside triphosphatase activity based on a malachite green ATPase assay. ST-610 is nonmutagenic, is well tolerated (nontoxic) in mice, and has shown efficacy in a sublethal murine model of DENV infection with the ability to significantly reduce viremia and viral load compared to vehicle controls.


Subject(s)
Benzoxazoles/therapeutic use , Dengue Virus/drug effects , Dengue Virus/enzymology , Dengue/drug therapy , RNA Helicases/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Virus Replication/drug effects , Animals , Cell Line , Dengue Virus/genetics , Dengue Virus/pathogenicity , Dogs , Drug Resistance, Viral/genetics , HeLa Cells , Humans , Mice , RNA Helicases/genetics , Vero Cells , Viral Nonstructural Proteins/genetics
15.
Antimicrob Agents Chemother ; 57(1): 15-25, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23070172

ABSTRACT

Dengue viruses (DENV) infect 50 to 100 million people worldwide per year, of which 500,000 develop severe life-threatening disease. This mosquito-borne illness is endemic in most tropical and subtropical countries and has spread significantly over the last decade. While there are several promising vaccine candidates in clinical trials, there are currently no approved vaccines or therapeutics available for treatment of dengue infection. Here, we describe a novel small-molecule compound, ST-148, that is a potent inhibitor of all four serotypes of DENV in vitro. ST-148 significantly reduced viremia and viral load in vital organs and tended to lower cytokine levels in the plasma in a nonlethal model of DENV infection in AG129 mice. Compound resistance mapped to the DENV capsid (C) gene, and a direct interaction of ST-148 with C protein is suggested by alterations of the intrinsic fluorescence of the protein in the presence of compound. Thus, ST-148 appears to interact with the DENV C protein and inhibits a distinct step(s) of the viral replication cycle.


Subject(s)
Antiviral Agents/pharmacology , Capsid Proteins/antagonists & inhibitors , Capsid Proteins/genetics , Dengue Virus/drug effects , Dengue/drug therapy , Heterocyclic Compounds, 3-Ring/pharmacology , Thiadiazoles/pharmacology , Viremia/drug therapy , Amino Acid Sequence , Animals , Antiviral Agents/chemical synthesis , Capsid Proteins/chemistry , Cell Line , Chlorocebus aethiops , Cricetinae , Dengue/virology , Dengue Virus/classification , Dengue Virus/genetics , Disease Models, Animal , Dogs , Escherichia coli/genetics , Heterocyclic Compounds, 3-Ring/chemical synthesis , Humans , Inhibitory Concentration 50 , Mice , Models, Molecular , Molecular Sequence Data , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Thiadiazoles/chemical synthesis , Viral Load/drug effects , Viral Plaque Assay , Viremia/virology , Virus Replication/drug effects
16.
J Virol ; 86(16): 8472-81, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22647699

ABSTRACT

The HIV-1 capsid (CA) protein plays essential roles in both early and late stages of virl replication and has emerged as a novel drug target. We report hybrid structure-based virtual screening to identify small molecules with the potential to interact with the N-terminal domain (NTD) of HIV-1 CA and disrupt early, preintegration steps of the HIV-1 replication cycle. The small molecule 4,4'-[dibenzo[b,d]furan-2,8-diylbis(5-phenyl-1H-imidazole-4,2-diyl)]dibenzoic acid (CK026), which had anti-HIV-1 activity in single- and multiple-round infections but failed to inhibit viral replication in peripheral blood mononuclear cells (PBMCs), was identified. Three analogues of CK026 with reduced size and better drug-like properties were synthesized and assessed. Compound I-XW-053 (4-(4,5-diphenyl-1H-imidazol-2-yl)benzoic acid) retained all of the antiviral activity of the parental compound and inhibited the replication of a diverse panel of primary HIV-1 isolates in PBMCs, while displaying no appreciable cytotoxicity. This antiviral activity was specific to HIV-1, as I-XW-053 displayed no effect on the replication of SIV or against a panel of nonretroviruses. Direct interaction of I-XW-053 was quantified with wild-type and mutant CA protein using surface plasmon resonance and isothermal titration calorimetry. Mutation of Ile37 and Arg173, which are required for interaction with compound I-XW-053, crippled the virus at an early, preintegration step. Using quantitative PCR, we demonstrated that treatment with I-XW-053 inhibited HIV-1 reverse transcription in multiple cell types, indirectly pointing to dysfunction in the uncoating process. In summary, we have identified a CA-specific compound that targets and inhibits a novel region in the NTD-NTD interface, affects uncoating, and possesses broad-spectrum anti-HIV-1 activity.


Subject(s)
Anti-HIV Agents/pharmacology , Capsid Proteins/antagonists & inhibitors , HIV-1/drug effects , HIV-1/physiology , Virus Uncoating/drug effects , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/toxicity , Calorimetry , Cell Line , Humans , Microbial Sensitivity Tests , Protein Binding , Real-Time Polymerase Chain Reaction , Reverse Transcription/drug effects , Simian Immunodeficiency Virus/drug effects , Surface Plasmon Resonance , Virus Internalization/drug effects , Virus Replication/drug effects
17.
J Biol Chem ; 287(30): 25203-15, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22645125

ABSTRACT

Staphylococcal enterotoxin B (SEB) is a potent toxin that can cause toxic shock syndrome and act as a lethal and incapacitating agent when used as a bioweapon. There are currently no vaccines or immunotherapeutics available against this toxin. Using phage display technology, human antigen-binding fragments (Fabs) were selected against SEB, and proteins were produced in Escherichia coli cells and characterized for their binding affinity and their toxin neutralizing activity in vitro and in vivo. Highly protective Fabs were converted into full-length IgGs and produced in mammalian cells. Additionally, the production of anti-SEB antibodies was explored in the Nicotiana benthamiana plant expression system. Affinity maturation was performed to produce optimized lead anti-SEB antibody candidates with subnanomolar affinities. IgGs produced in N. benthamiana showed characteristics comparable with those of counterparts produced in mammalian cells. IgGs were tested for their therapeutic efficacy in the mouse toxic shock model using different challenge doses of SEB and a treatment with 200 µg of IgGs 1 h after SEB challenge. The lead candidates displayed full protection from lethal challenge over a wide range of SEB challenge doses. Furthermore, mice that were treated with anti-SEB IgG had significantly lower IFNγ and IL-2 levels in serum compared with mock-treated mice. In summary, these anti-SEB monoclonal antibodies represent excellent therapeutic candidates for further preclinical and clinical development.


Subject(s)
Antibodies, Monoclonal/pharmacology , Enterotoxins/antagonists & inhibitors , Immunoglobulin Fab Fragments/pharmacology , Shock, Septic/therapy , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibody Affinity/genetics , Enterotoxins/immunology , Enterotoxins/toxicity , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Mice , Mice, Inbred BALB C , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Shock, Septic/chemically induced , Shock, Septic/genetics , Shock, Septic/immunology , Time Factors , Nicotiana/genetics
18.
PLoS One ; 7(4): e34761, 2012.
Article in English | MEDLINE | ID: mdl-22496857

ABSTRACT

Alphaviruses, including Venezuelan Equine Encephalitis Virus (VEEV), cause disease in both equine and humans that exhibit overt encephalitis in a significant percentage of cases. Features of the host immune response and tissue-specific responses may contribute to fatal outcomes as well as the development of encephalitis. It has previously been shown that VEEV infection of mice induces transcription of pro-inflammatory cytokines genes (e.g., IFN-γ, IL-6, IL-12, iNOS and TNF-α) within 6 h. GSK-3ß is a host protein that is known to modulate pro-inflammatory gene expression and has been a therapeutic target in neurodegenerative disorders such as Alzheimer's. Hence inhibition of GSK-3ß in the context of encephalitic viral infections has been useful in a neuroprotective capacity. Small molecule GSK-3ß inhibitors and GSK-3ß siRNA experiments indicated that GSK-3ß was important for VEEV replication. Thirty-eight second generation BIO derivatives were tested and BIOder was found to be the most potent inhibitor, with an IC(50) of ∼0.5 µM and a CC(50) of >100 µM. BIOder was a more potent inhibitor of GSK-3ß than BIO, as demonstrated through in vitro kinase assays from uninfected and infected cells. Size exclusion chromatography experiments demonstrated that GSK-3ß is found in three distinct complexes in VEEV infected cells, whereas GSK-3ß is only present in one complex in uninfected cells. Cells treated with BIOder demonstrated an increase in the anti-apoptotic gene, survivin, and a decrease in the pro-apoptotic gene, BID, suggesting that modulation of pro- and anti-apoptotic genes contributes to the protective effect of BIOder treatment. Finally, BIOder partially protected mice from VEEV induced mortality. Our studies demonstrate the utility of GSK-3ß inhibitors for modulating VEEV infection.


Subject(s)
Encephalitis Virus, Venezuelan Equine/drug effects , Encephalomyelitis, Venezuelan Equine/drug therapy , Enzyme Inhibitors/therapeutic use , Glycogen Synthase Kinase 3/antagonists & inhibitors , Animals , BH3 Interacting Domain Death Agonist Protein/analysis , Encephalomyelitis, Venezuelan Equine/mortality , Female , Glycogen Synthase Kinase 3 beta , Inhibitor of Apoptosis Proteins/analysis , Mice , Mice, Inbred C3H , Repressor Proteins/analysis , Survivin , Virus Replication/drug effects
19.
Antiviral Res ; 83(3): 245-51, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19523489

ABSTRACT

We report herein the identification of a small molecule therapeutic, FGI-106, which displays potent and broad-spectrum inhibition of lethal viral hemorrhagic fevers pathogens, including Ebola, Rift Valley and Dengue Fever viruses, in cell-based assays. Using mouse models of Ebola virus, we further demonstrate that FGI-106 can protect animals from an otherwise lethal infection when used either in a prophylactic or therapeutic setting. A single treatment, administered 1 day after infection, is sufficient to protect animals from lethal Ebola virus challenge. Cell-based assays also identified inhibitory activity against divergent virus families, which supports a hypothesis that FGI-106 interferes with a common pathway utilized by different viruses. These findings suggest FGI-106 may provide an opportunity for targeting viral diseases.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Ebolavirus/drug effects , Animals , Cell Line, Tumor , Cell Survival , Chemoprevention , Dengue Virus/drug effects , Female , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/prevention & control , Male , Mice , Mice, Inbred BALB C , Molecular Structure , Rift Valley fever virus/drug effects , Viral Plaque Assay
SELECTION OF CITATIONS
SEARCH DETAIL
...