Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Virol ; 98(4): e0013224, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38511932

ABSTRACT

Heartland virus (HRTV) is an emerging tick-borne bandavirus that causes a febrile illness of varying severity in humans, with cases reported in eastern and midwestern regions of the United States. No vaccines or approved therapies are available to prevent or treat HRTV disease. Here, we describe the genetic changes, natural history of disease, and pathogenesis of a mouse-adapted HRTV (MA-HRTV) that is uniformly lethal in 7- to 8-week-old AG129 mice at low challenge doses. We used this model to assess the efficacy of the ribonucleoside analog, 4'-fluorouridine (EIDD-2749), and showed that once-daily oral treatment with 3 mg/kg of drug, initiated after the onset of disease, protects mice against lethal MA-HRTV challenge and reduces viral loads in blood and tissues. Our findings provide insights into HRTV virulence and pathogenesis and support further development of EIDD-2749 as a therapeutic intervention for HRTV disease. IMPORTANCE: More than 60 cases of HRTV disease spanning 14 states have been reported to the United States Centers for Disease Control and Prevention. The expanding range of the Lone Star tick that transmits HRTV, the growing population of at-risk persons living in geographic areas where the tick is abundant, and the lack of antiviral treatments or vaccines raise significant public health concerns. Here, we report the development of a new small-animal model of lethal HRTV disease to gain insight into HRTV pathogenesis and the application of this model for the preclinical development of a promising new antiviral drug candidate, EIDD-2749. Our findings shed light on how the virus causes disease and support the continued development of EIDD-2749 as a therapeutic for severe cases of HRTV infection.


Subject(s)
Bunyaviridae Infections , Bunyaviridae , Uracil Nucleotides , Animals , Humans , Mice , Bunyaviridae Infections/drug therapy , Ticks , United States , Uracil Nucleotides/therapeutic use
2.
J Virol ; 98(4): e0011224, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38506509

ABSTRACT

Live-attenuated virus vaccines provide long-lived protection against viral disease but carry inherent risks of residual pathogenicity and genetic reversion. The live-attenuated Candid#1 vaccine was developed to protect Argentines against lethal infection by the Argentine hemorrhagic fever arenavirus, Junín virus. Despite its safety and efficacy in Phase III clinical study, the vaccine is not licensed in the US, in part due to concerns regarding the genetic stability of attenuation. Previous studies had identified a single F427I mutation in the transmembrane domain of the Candid#1 envelope glycoprotein GPC as the key determinant of attenuation, as well as the propensity of this mutation to revert upon passage in cell culture and neonatal mice. To ascertain the consequences of this reversion event, we introduced the I427F mutation into recombinant Candid#1 (I427F rCan) and investigated the effects in two validated small-animal models: in mice expressing the essential virus receptor (human transferrin receptor 1; huTfR1) and in the conventional guinea pig model. We report that I427F rCan displays only modest virulence in huTfR1 mice and appears attenuated in guinea pigs. Reversion at another attenuating locus in Candid#1 GPC (T168A) was also examined, and a similar pattern was observed. By contrast, virus bearing both revertant mutations (A168T+I427F rCan) approached the lethal virulence of the pathogenic Romero strain in huTfR1 mice. Virulence was less extreme in guinea pigs. Our findings suggest that genetic stabilization at both positions is required to minimize the likelihood of reversion to virulence in a second-generation Candid#1 vaccine.IMPORTANCELive-attenuated virus vaccines, such as measles/mumps/rubella and oral poliovirus, provide robust protection against disease but carry with them the risk of genetic reversion to the virulent form. Here, we analyze the genetics of reversion in the live-attenuated Candid#1 vaccine that is used to protect against Argentine hemorrhagic fever, an often-lethal disease caused by the Junín arenavirus. In two validated small-animal models, we find that restoration of virulence in recombinant Candid#1 viruses requires back-mutation at two positions specific to the Candid#1 envelope glycoprotein GPC, at positions 168 and 427. Viruses bearing only a single change showed only modest virulence. We discuss strategies to genetically harden Candid#1 GPC against these two reversion events in order to develop a safer second-generation Candid#1 vaccine virus.


Subject(s)
Hemorrhagic Fever, American , Junin virus , Viral Vaccines , Animals , Guinea Pigs , Humans , Mice , Glycoproteins/genetics , Hemorrhagic Fever, American/prevention & control , Junin virus/physiology , South American People , Vaccines, Attenuated/genetics , Viral Vaccines/genetics , Virulence
3.
Article in English | MEDLINE | ID: mdl-33558299

ABSTRACT

The impact of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of COVID-19, is global and unprecedented. Although remdesivir has recently been approved by the FDA to treat SARS-CoV-2 infection, no oral antiviral is available for outpatient treatment. AT-527, an orally administered double prodrug of a guanosine nucleotide analog, was previously shown to be highly efficacious and well tolerated in hepatitis C virus (HCV)-infected subjects. Here, we report the potent in vitro activity of AT-511, the free base of AT-527, against several coronaviruses, including SARS-CoV-2. In normal human airway epithelial cells, the concentration of AT-511 required to inhibit replication of SARS-CoV-2 by 90% (EC90) was 0.47 µM, very similar to its EC90 against human coronavirus (HCoV)-229E, HCoV-OC43, and SARS-CoV in Huh-7 cells. Little to no cytotoxicity was observed for AT-511 at concentrations up to 100 µM. Substantial levels of the active triphosphate metabolite AT-9010 were formed in normal human bronchial and nasal epithelial cells incubated with 10 µM AT-511 (698 ± 15 and 236 ± 14 µM, respectively), with a half-life of at least 38 h. Results from steady-state pharmacokinetic and tissue distribution studies of nonhuman primates administered oral doses of AT-527, as well as pharmacokinetic data from subjects given daily oral doses of AT-527, predict that twice daily oral doses of 550 mg AT-527 will produce AT-9010 trough concentrations in human lung that exceed the EC90 observed for the prodrug against SARS-CoV-2 replication. This suggests that AT-527 may be an effective treatment option for COVID-19.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Guanosine Monophosphate/analogs & derivatives , Guanosine/pharmacology , Phosphoramides/pharmacology , Prodrugs/pharmacology , SARS-CoV-2/drug effects , Administration, Oral , Animals , COVID-19/virology , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Coronavirus 229E, Human/metabolism , Coronavirus OC43, Human/metabolism , Cricetinae , Epithelial Cells/virology , Guanosine Monophosphate/pharmacology , Humans , Lung/virology , SARS-CoV-2/metabolism , Vero Cells , Virus Replication/drug effects
4.
Cell Cycle ; 19(24): 3632-3638, 2020 12.
Article in English | MEDLINE | ID: mdl-33305659

ABSTRACT

PT150 is a clinical-stage molecule, taken orally, with a strong safety profile having completed Phase 1 and Phase 2 clinical trials for its original use as an antidepressant. It has an active IND for COVID-19. Antiviral activities have been found for PT150 and other members of its class in a variety of virus families; thus, it was now tested against SARS-CoV-2 in human bronchial epithelial lining cells and showed effective 90% inhibitory antiviral concentration (EC90) of 5.55 µM. PT150 is a member of an extended platform of novel glucocorticoid receptor (GR) and androgen receptor (AR) modulating molecules. In vivo, their predominant net effect is one of systemic glucocorticoid antagonism, but they also show direct downregulation of AR and minor GR agonism at the cellular level. We hypothesize that anti-SARS-CoV-2 activity depends in part on this AR downregulation through diminished TMPRSS2 expression and modulation of ACE2 activity. Given that hypercortisolemia is now suggested to be a significant co-factor for COVID-19 progression, we also postulate an additive role for its potent immunomodulatory effects through systemic antagonism of cortisol.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Receptors, Androgen/metabolism , Receptors, Glucocorticoid/metabolism , SARS-CoV-2/drug effects , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/virology , Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/therapeutic use , Cell Line , Disease Progression , Down-Regulation , Glucocorticoids/antagonists & inhibitors , Glucocorticoids/metabolism , Humans , Hydrocortisone/antagonists & inhibitors , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Middle East Respiratory Syndrome Coronavirus/drug effects , Receptors, Glucocorticoid/agonists , Serine Endopeptidases/metabolism
5.
PLoS Pathog ; 14(12): e1007439, 2018 12.
Article in English | MEDLINE | ID: mdl-30576397

ABSTRACT

Arenaviruses are a significant cause of hemorrhagic fever, an often-fatal disease for which there is no approved antiviral therapy. Lassa fever in particular generates high morbidity and mortality in West Africa, where the disease is endemic, and a recent outbreak in Nigeria was larger and more geographically diverse than usual. We are developing LHF-535, a small-molecule viral entry inhibitor that targets the arenavirus envelope glycoprotein, as a therapeutic candidate for Lassa fever and other hemorrhagic fevers of arenavirus origin. Using a lentiviral pseudotype infectivity assay, we determined that LHF-535 had sub-nanomolar potency against the viral envelope glycoproteins from all Lassa virus lineages, with the exception of the glycoprotein from the LP strain from lineage I, which was 100-fold less sensitive than that of other strains. This reduced sensitivity was mediated by a unique amino acid substitution, V434I, in the transmembrane domain of the envelope glycoprotein GP2 subunit. This position corresponds to the attenuation determinant of Candid#1, a live-attenuated Junín virus vaccine strain used to prevent Argentine hemorrhagic fever. Using a virus-yield reduction assay, we determined that LHF-535 potently inhibited Junín virus, but not Candid#1, and the Candid#1 attenuation determinant, F427I, regulated this difference in sensitivity. We also demonstrated that a daily oral dose of LHF-535 at 10 mg/kg protected mice from a lethal dose of Tacaribe virus. Serial passage of Tacaribe virus in LHF-535-treated Vero cells yielded viruses that were resistant to LHF-535, and the majority of drug-resistant viruses exhibited attenuated pathogenesis. These findings provide a framework for the clinical development of LHF-535 as a broad-spectrum inhibitor of arenavirus entry and provide an important context for monitoring the emergence of drug-resistant viruses.


Subject(s)
Antiviral Agents/pharmacology , Lassa Fever , Lassa virus/genetics , Virulence/drug effects , Virulence/genetics , Animals , Chlorocebus aethiops , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , HEK293 Cells , Humans , Lassa virus/drug effects , Mice , Mutation , Vero Cells , Viral Envelope Proteins/genetics
6.
Antiviral Res ; 160: 48-54, 2018 12.
Article in English | MEDLINE | ID: mdl-30339848

ABSTRACT

2'-Fluoro-2'-deoxycytidine (2'-FdC) was reported to inhibit various viruses in vitro, including Borna disease, hepatitis C, Lassa fever, influenza and certain herpes viruses, and is inhibitory to influenza viruses in mice. We investigated the antiviral activity of 2'-FdC against several unrelated bunyaviruses in 50% cytopathic effect (CPE) inhibition assays and, with viruses that cause limited CPE, 90% virus yield reduction (VYR) assays. La Crosse (LACV), Maporal, Punta Toro, Rift Valley fever (RVFV), and San Angelo viruses were inhibited in CPE assays at 2.2-9.7 µM concentrations. In VYR assays, Heartland and severe fever with thrombocytopenia syndrome (SFTSV) viruses were inhibited at 0.9 and 3.7 µM, respectively. In contrast, ribavirin inhibited these viruses at an average of 47 µM. Antiviral efficacy studies were also conducted in mice infected with RVFV, SFTSV, and LACV. Against RVFV, 2'-FdC (100 and 200 mg/kg/day) and ribavirin (100 mg/kg/day) treatments each delayed mortality by approximately 6 days compared to placebo. Liver, spleen, and serum viral titers were significantly reduced by antiviral treatments. 2'-FdC (100 and 200 mg/kg/day) prevented death in SFTSV-infected mice, but was not as effective as favipiravir (100 mg/kg/day) based on body weight loss during infection. The 100 mg/kg/day doses of 2'-FdC and favipiravir significantly reduced liver, spleen, and serum viral titers. 2'-FdC and ribavirin afforded no protection against LACV infection in mice, which is encephalitic and thus inherently more difficult to treat. Taken together, our data suggest that 2'-FdC may be a viable candidate for treating certain non-encephalitic bunyavirus infections such as those caused by phleboviruses.


Subject(s)
Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Bunyaviridae Infections/drug therapy , DNA Viruses/drug effects , Deoxycytidine/analogs & derivatives , RNA Viruses/drug effects , Animal Structures/virology , Animals , Body Weight , Cytopathogenic Effect, Viral , DNA Viruses/growth & development , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Disease Models, Animal , Mice , Microbial Sensitivity Tests , Placebos/administration & dosage , RNA Viruses/growth & development , Survival Analysis , Treatment Outcome , Viral Load
7.
Antiviral Res ; 156: 38-45, 2018 08.
Article in English | MEDLINE | ID: mdl-29864447

ABSTRACT

Rift Valley fever virus (RVFV) is a mosquito-borne pathogen endemic to sub-Saharan Africa and the Arabian Peninsula. There are no approved antiviral therapies or vaccines available to treat or prevent severe disease associated with RVFV infection in humans. The adenosine analog, galidesivir (BCX4430), is a broad-spectrum antiviral drug candidate with in vitro antiviral potency (EC50 of less than 50 µM) in more than 20 different viruses across eight different virus families. Here we report on the activity of galidesivir in the hamster model of peracute RVFV infection. Intramuscular and intraperitoneal treatments effectively limited systemic RVFV (strain ZH501) infection as demonstrated by significantly improved survival outcomes and the absence of infectious virus in the spleen and the majority of the serum, brain, and liver samples collected from infected animals. Our findings support the further development of galidesivir as an antiviral therapy for use in treating severe RVFV infection, and possibly other related phleboviral diseases.


Subject(s)
Antiviral Agents/administration & dosage , Purine Nucleosides/administration & dosage , Rift Valley Fever/drug therapy , Rift Valley fever virus/drug effects , Adenine/analogs & derivatives , Adenosine/analogs & derivatives , Animals , Disease Models, Animal , Injections, Intramuscular , Injections, Intraperitoneal , Liver/virology , Mesocricetus , Pyrrolidines , Spleen/virology , Survival Analysis , Treatment Outcome
8.
J Infect Dis ; 218(4): 522-527, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29762684

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV) poses a substantial risk to immunocompromised individuals. The case fatality rate in recent clusters of LCMV infection in immunosuppressed organ transplantation recipients has exceeded 70%. In the present study, we demonstrate potent antiviral activity of favipiravir against acute, disseminated LCMV infection in NZB mice. Treatment resulted in complete protection against mortality and dramatic reductions in viral loads. In contrast, ribavirin, the current antiviral of choice, was mostly ineffective. Our findings, and the high lethality associated with LCMV infection in transplant recipients, support the consideration of favipiravir as a first-line therapeutic option.


Subject(s)
Amides/administration & dosage , Antiviral Agents/administration & dosage , Lymphocytic Choriomeningitis/drug therapy , Lymphocytic choriomeningitis virus/isolation & purification , Pyrazines/administration & dosage , Viral Load , Animals , Disease Models, Animal , Female , Immunocompromised Host , Lymphocytic Choriomeningitis/virology , Male , Mice, Inbred NZB , Ribavirin/administration & dosage , Survival Analysis , Transplant Recipients , Treatment Outcome
9.
Virology ; 511: 175-183, 2017 11.
Article in English | MEDLINE | ID: mdl-28865344

ABSTRACT

Heartland virus (HRTV) is an emerging tick-borne virus (Bunyaviridae, Phlebovirus) that has caused sporadic cases of human disease in several central and mid-eastern states of America. Animal models of HRTV disease are needed to gain insights into viral pathogenesis and advancing antiviral drug development. Presence of clinical disease following HRTV challenge in hamsters deficient in STAT2 function underscores the important role played by type I interferon-induced antiviral responses. However, the recovery of most of the infected animals suggests that other mechanisms to control infection and limit disease offer substantial protection. The most prominent disease sign with HRTV infection in STAT2 knockout hamsters was dramatic weight loss with clinical laboratory and histopathology demonstrating acute inflammation in the spleen, lymph node, liver and lung. Finally, we show that HRTV disease in hamsters can be prevented by the use of favipiravir, a promising broad-spectrum antiviral in clinical development for the treatment of influenza.


Subject(s)
Amides/therapeutic use , Antiviral Agents/therapeutic use , Bunyaviridae Infections/pathology , Bunyaviridae Infections/prevention & control , Pyrazines/therapeutic use , STAT2 Transcription Factor/deficiency , Animal Structures/pathology , Animals , Chemoprevention , Cricetinae , Disease Models, Animal , Inflammation/pathology , Interferon Type I/immunology , Treatment Outcome
10.
J Virol Methods ; 246: 51-57, 2017 08.
Article in English | MEDLINE | ID: mdl-28359770

ABSTRACT

Studies were conducted to determine the performance of four dyes in assessing antiviral activities of compounds against three RNA viruses with differing cytopathogenic properties. Dyes included alamarBlue® measured by absorbance (ALB-A) and fluorescence (ALB-F), neutral red (NR), Viral ToxGlo™ (VTG), and WST-1. Viruses were chikungunya, dengue type 2, and Junin, which generally cause 100, 80-90, and 50% maximal cytopathic effect (CPE), respectively, in Vero or Vero 76 cells Compounds evaluated were 6-azauridine, BCX-4430, 3-deazaguanine, EICAR, favipiravir, infergen, mycophenolic acid (MPA), ribavirin, and tiazofurin. The 50% virus-inhibitory (EC50) values for each inhibitor and virus combination did not vary significantly based on the dye used. However, dyes varied in distinguishing the vitality of virus-infected cultures when not all cells were killed by virus infection. For example, VTG uptake into dengue-infected cells was nearly 50% when visual examination showed only 10-20% cell survival. ALB-A measured infected cell viability differently than ALB-F as follows: 16% versus 32% (dengue-infected), respectively, and 51% versus 72% (Junin-infected), respectively. Cytotoxicity (CC50) assays with dyes in uninfected proliferating cells produced similar CC50 values for EICAR (1.5-8.9µM) and MPA (0.8-2.5µM). 6-Azauridine toxicity was 6.1-17.5µM with NR, VTG, and WST-1, compared to 48-92µM with ALB-A and ALB-F (P<0.001). Curiously, the CC50 values for 3-deazaguanine were 83-93µM with ALB-F versus 2.4-7.0µM with all other dyes including ALB-A (P<0.001). Overall, ALB minimized the toxicities detected with these two inhibitors. Because the choice of dyes affected CC50 values, this impacted on the resulting in vitro selectivity indexes (calculated as CC50/EC50 ratio).


Subject(s)
Antiviral Agents/pharmacology , Cell Survival/drug effects , Coloring Agents , Cytopathogenic Effect, Viral , RNA Viruses/drug effects , Viruses/drug effects , Animals , Chikungunya virus/drug effects , Chikungunya virus/pathogenicity , Chikungunya virus/physiology , Chlorocebus aethiops , Coloring Agents/chemistry , Dengue Virus/drug effects , Dengue Virus/pathogenicity , Dengue Virus/physiology , Junin virus/drug effects , Junin virus/pathogenicity , Junin virus/physiology , Oxazines , RNA Viruses/pathogenicity , RNA Viruses/physiology , Vero Cells , Virus Replication/drug effects , Xanthenes
11.
J Virol ; 91(3)2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27881648

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne disease endemic in parts of Asia. The etiologic agent, SFTS virus (SFTSV; family Bunyaviridae, genus Phlebovirus) has caused significant morbidity and mortality in China, South Korea, and Japan, with key features of disease being intense fever, thrombocytopenia, and leukopenia. Case fatality rates are estimated to be in the 30% range, and no antivirals or vaccines are approved for use for treatment and prevention of SFTS. There is evidence that in human cells, SFTSV sequesters STAT proteins in replication complexes, thereby inhibiting type I interferon signaling. Here, we demonstrate that hamsters devoid of functional STAT2 are highly susceptible to as few as 10 PFU of SFTSV, with animals generally succumbing within 5 to 6 days after subcutaneous challenge. The disease included marked thrombocytopenia and inflammatory disease characteristic of the condition in humans. Infectious virus titers were present in the blood and most tissues 3 days after virus challenge, and severe inflammatory lesions were found in the spleen and liver samples of SFTSV-infected hamsters. We also show that SFTSV infection in STAT2 knockout (KO) hamsters is responsive to favipiravir treatment, which protected all animals from lethal disease and reduced serum and tissue viral loads by 3 to 6 orders of magnitude. Taken together, our results provide additional insights into the pathogenesis of SFTSV infection and support the use of the newly described STAT2 KO hamster model for evaluation of promising antiviral therapies. IMPORTANCE: Severe fever with thrombocytopenia syndrome (SFTS) is an emerging viral disease for which there are currently no therapeutic options or available vaccines. The causative agent, SFTS virus (SFTSV), is present in China, South Korea, and Japan, and infections requiring medical attention result in death in as many as 30% of the cases. Here, we describe a novel model of SFTS in hamsters genetically engineered to be deficient in a protein that helps protect humans and animals against viral infections. These hamsters were found to be susceptible to SFTSV and share disease features associated with the disease in humans. Importantly, we also show that SFTSV infection in hamsters can be effectively treated with a broad-spectrum antiviral drug approved for use in Japan. Our findings suggest that the new SFTS model will be an excellent resource to better understand SFTSV infection and disease as well as a valuable tool for evaluating promising antiviral drugs.


Subject(s)
Bunyaviridae Infections/virology , Models, Biological , Phlebovirus/physiology , Amides/pharmacology , Animals , Animals, Genetically Modified , Antiviral Agents/pharmacology , Bunyaviridae Infections/drug therapy , Bunyaviridae Infections/genetics , Bunyaviridae Infections/mortality , Cricetinae , Disease Models, Animal , Disease Susceptibility , Genotype , Humans , Phenotype , Pyrazines/pharmacology , STAT2 Transcription Factor/genetics
12.
Antiviral Res ; 126: 62-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26711718

ABSTRACT

Favipiravir is approved in Japan to treat novel or re-emerging influenza viruses, and is active against a broad spectrum of RNA viruses, including Ebola. Ribavirin is the only other licensed drug with activity against multiple RNA viruses. Recent studies show that ribavirin and favipiravir act synergistically to inhibit bunyavirus infections in cultured cells and laboratory mice, likely due to their different mechanisms of action. Convalescent immune globulin is the only approved treatment for Argentine hemorrhagic fever caused by the rodent-borne Junin arenavirus. We previously reported that favipiravir is highly effective in a number of small animal models of Argentine hemorrhagic fever. We now report that addition of low dose of ribavirin synergistically potentiates the activity of favipiravir against Junin virus infection of guinea pigs and another arenavirus, Pichinde virus infection of hamsters. This suggests that the efficacy of favipiravir against hemorrhagic fever viruses can be further enhanced through the addition of low-dose ribavirin.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Hemorrhagic Fevers, Viral/drug therapy , Pyrazines/pharmacology , RNA Viruses/drug effects , Ribavirin/pharmacology , Animals , Arenavirus/drug effects , Chlorocebus aethiops , Cricetinae , Dengue Virus/drug effects , Disease Models, Animal , Drug Synergism , Female , Guinea Pigs , Orthohantavirus/drug effects , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever, American/drug therapy , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fevers, Viral/blood , Hemorrhagic Fevers, Viral/veterinary , Hemorrhagic Fevers, Viral/virology , Junin virus/drug effects , Male , Mesocricetus , Mice , Vero Cells
13.
Front Microbiol ; 6: 651, 2015.
Article in English | MEDLINE | ID: mdl-26175722

ABSTRACT

Rift Valley fever virus (RVFV; Bunyaviridae, Phlebovirus) causes a range of illnesses that include retinitis, fulminant hepatitis, neurologic disease, and hemorrhagic fever. In hospitalized individuals, case fatality rates can be as high as 10-20%. There are no vaccines or antivirals approved for human use to prevent or treat severe RVFV infections. We previously tested the efficacy of the MP-12 vaccine strain and related variants with NSs truncations as a post-exposure prophylaxis in mice infected with wild-type pathogenic RVFV strain ZH501. Post-exposure efficacy of the rMP12-C13type, a recombinant MP-12 vaccine virus which encodes an in-frame truncation removing 69% of the NSs protein, resulted in 30% survival when administering the virus within 30 min of subcutaneous ZH501 challenge in mice, while the parental MP-12 virus conferred no protection by post-exposure vaccination. Here, we demonstrate uniform protection of hamsters by post-exposure vaccination with rMP12-C13type administered 6 h post-ZH501 infection while no efficacy was observed with the parental MP-12 virus. Notably, both the MP-12 and rMP12-C13type viruses were highly effective (100% protection) when administered 21 days prior to challenge. In a subsequent study delaying vaccination until 8, 12, and 24 h post-RVFV exposure, we observed 80, 70, and 30% survival, respectively. Our findings indicate that the rapid protective innate immune response elicited by rMP12-C13type may be due to the truncated NSs protein, suggesting that the resulting functional inactivation of NSs plays an important role in the observed post-exposure efficacy. Taken together, the data demonstrate that post-exposure vaccination with rMP12-C13type is effective in limiting ZH501 replication and associated disease in standard pre-exposure vaccination and post-challenge treatment models of RVFV infection, and suggest an extended post-exposure prophylaxis window beyond that initially observed in mice.

14.
Antivir Chem Chemother ; 23(4): 151-9, 2014 Jan 29.
Article in English | MEDLINE | ID: mdl-23337126

ABSTRACT

BACKGROUND: Junín virus (JUNV) and several other clade B New World arenaviruses cause human disease ranging from mild febrile illness to severe viral haemorrhagic fever. These viruses pose a significant threat to national security and safe and effective therapies are limited except in Argentina, where immune plasma is the standard of care for treating JUNV infection in cases of Argentine haemorrhagic fever. METHODS: An in vitro screen of the Chemtura library identified several compounds with activity against Tacaribe virus (TCRV), a clade B arenavirus closely related to JUNV. Of these compounds, D746, a phenolic dibenzylsulfide, was further pursued for additional in vitro studies and evaluated in the AG129 mouse TCRV infection model. RESULTS: D746 was found to act during an early to intermediate stage of the TCRV replication cycle and µM range activity was confirmed by virus yield reduction assays with both TCRV and JUNV. Although intraperitoneal twice daily treatment regimens were found to be highly effective when started 2 h prior to TCRV challenge in AG129 mice, post-exposure treatment initiated 3 days after infection was not efficacious. Interestingly, despite the pre-exposure treatment success, D746 did not reduce serum or tissue virus titres during the acute infection. Moreover, D746 elicited ascites fluid accumulation in mice during, as well as independent of, infection. CONCLUSIONS: Our findings suggest that D746 may be altering the host response to TCRV infection in AG129 mice in a way that limits pathogenesis and thereby protects mice from otherwise lethal infection in the absence of measurable reductions in viral burden.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Arenaviruses, New World/drug effects , Hemorrhagic Fever, American/drug therapy , Animals , Chlorocebus aethiops , Humans , Junin virus/drug effects , Mice , Phenols/chemistry , Phenols/therapeutic use , Sulfides/chemistry , Sulfides/therapeutic use , Vero Cells
15.
Antimicrob Agents Chemother ; 56(8): 4168-74, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22615273

ABSTRACT

Punta Toro virus (PTV; Bunyaviridae, Phlebovirus) is related to Rift Valley fever virus (RVFV), a pathogenic agent which causes severe disease in humans and livestock primarily in the sub-Saharan region of Africa. The recent range expansion of RVFV and the potential for its intentional release into naïve populations pose a significant threat to public health and agriculture. Studies modeling disease in rodents and nonhuman primates have shown that PTV and RVFV are highly sensitive to the antiviral effects of alpha interferon (IFN-α), an important component of the innate antiviral host response. While recombinant IFN-α has high therapeutic value, its utility for the treatment of neglected tropical diseases is hindered by its short in vivo half-life and costly production of longer-lasting pegylated IFNs. Here, we demonstrate extended preexposure protection against lethal PTV challenge following a single intranasal administration of DEF201, which is a replication-deficient human adenovirus type 5 vector engineered to constitutively express consensus IFN-α (cIFN-α) from transduced host cells. DEF201 was also efficacious when administered within 24 h as a postexposure countermeasure. Serum concentrations of cIFN-α could be detected as early as 8 h following treatment and persisted for more than 1 week. The prolonged antiphlebovirus prophylactic effect, low production costs, and ease of administration make DEF201 a promising agent for intervention during natural disease outbreaks and for countering possible bioterrorist acts.


Subject(s)
Adenoviridae/genetics , Bunyaviridae Infections/prevention & control , Interferon-alpha/genetics , Interferon-alpha/metabolism , Phlebovirus , Administration, Intranasal , Animals , Antiviral Agents/blood , Antiviral Agents/metabolism , Cricetinae , Female , Genetic Vectors , Interferon-alpha/blood , Liver/virology , Mesocricetus , Recombinant Proteins
16.
Antivir Chem Chemother ; 21(5): 193-200, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21566265

ABSTRACT

BACKGROUND: Pathogenic hantaviruses geographically distributed in the Old World cause haemorrhagic fever with renal syndrome (HFRS), whereas New World hantaviruses are the aetiological agents of hantavirus cardiopulmonary syndrome (HCPS). Ribavirin, a drug associated with toxicities, is presently indicated for treatment of HFRS, whereas treatment of the more frequently lethal HCPS is limited to supportive care. Because of the need for safe and effective antivirals to treat severe hantaviral infections, we evaluated favipiravir (T-705) against Dobrava and Maporal viruses as representative Old World and New World hantaviruses, respectively. Dobrava virus causes HFRS in Europe. Maporal virus (MPRLV), recently isolated from western Venezuela, is phylogenetically similar to Andes virus, the principal cause of HCPS in Argentina. METHODS: Hantavirus replication in the presence of various inhibitors was measured by focus-forming unit assays and quantitative reverse transcriptase PCR. Phylogenetic relationships were assessed by the neighbour-joining and bootstrap consensus methods. RESULTS: Here, we show that infection of Vero E6 cells with MPRLV is dependent on ß3 integrins, similar to that reported for pathogenic hantaviruses. Furthermore, by analysis of molecular determinants associated with the G1 glycoprotein cytoplasmic tail, we show the close genetic proximity of MPRLV to other HCPS-causing hantaviruses in these regions predictive of pathogenicity. We also demonstrate anti-hantavirus activity by favipiravir with inhibitory concentrations ranging from 65 to 93 µM and selectivity indices>50. CONCLUSIONS: Our data suggest that MPRLV may serve as a safer alternative to modelling infection caused by the highly lethal Andes virus and that hantaviruses are sensitive to the effects of favipiravir in cell culture.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Orthohantavirus/drug effects , Orthohantavirus/genetics , Pyrazines/pharmacology , Animals , Cells, Cultured , Chlorocebus aethiops , Orthohantavirus/pathogenicity , Microbial Sensitivity Tests , Vero Cells , Virus Replication/drug effects
17.
PLoS One ; 5(9)2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20862280

ABSTRACT

BACKGROUND: A growing number of arenaviruses can cause a devastating viral hemorrhagic fever (VHF) syndrome. They pose a public health threat as emerging viruses and because of their potential use as bioterror agents. All of the highly pathogenic New World arenaviruses (NWA) phylogenetically segregate into clade B and require maximum biosafety containment facilities for their study. Tacaribe virus (TCRV) is a nonpathogenic member of clade B that is closely related to the VHF arenaviruses at the amino acid level. Despite this relatedness, TCRV lacks the ability to antagonize the host interferon (IFN) response, which likely contributes to its inability to cause disease in animals other than newborn mice. METHODOLOGY/PRINCIPAL FINDINGS: Here we describe a new mouse model based on TCRV challenge of AG129 IFN-α/ß and -γ receptor-deficient mice. Titration of the virus by intraperitoneal (i.p.) challenge of AG129 mice resulted in an LD(50) of ∼100 fifty percent cell culture infectious doses. Virus replication was evident in the serum, liver, lung, spleen, and brain 4-8 days after inoculation. MY-24, an aristeromycin derivative active against TCRV in cell culture at 0.9 µM, administered i.p. once daily for 7 days, offered highly significant (P<0.001) protection against mortality in the AG129 mouse TCRV infection model, without appreciably reducing viral burden. In contrast, in a hamster model of arenaviral hemorrhagic fever based on challenge with clade A Pichinde arenavirus, MY-24 did not offer significant protection against mortality. CONCLUSIONS/SIGNIFICANCE: MY-24 is believed to act as an inhibitor of S-adenosyl-L-homocysteine hydrolase, but our findings suggest that it may ameliorate disease by blunting the effects of the host response that play a role in disease pathogenesis. The new AG129 mouse TCRV infection model provides a safe and cost-effective means to conduct early-stage pre-clinical evaluations of candidate antiviral therapies that target clade B arenaviruses.


Subject(s)
Adenosine/analogs & derivatives , Antiviral Agents/administration & dosage , Arenaviridae Infections/drug therapy , Arenaviruses, New World/physiology , Disease Models, Animal , Mice , Adenosine/administration & dosage , Adenosine/chemistry , Animals , Antiviral Agents/chemistry , Arenaviridae Infections/mortality , Arenaviridae Infections/virology , Arenaviruses, New World/drug effects , Chlorocebus aethiops , Cricetinae , Female , Humans , Male , Mesocricetus , Mice, Knockout , Vero Cells , Virus Replication
18.
Antiviral Res ; 86(3): 261-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20227442

ABSTRACT

Yellow fever virus (YFV) continues to cause outbreaks of disease in endemic areas where vaccine is underutilized. Due to the effectiveness of the vaccine, antiviral development solely for the treatment of YFV is not feasible, but antivirals that are effective in the treatment of related viral diseases may be characterized for potential use against YFV as a secondary indication disease. 2'-C-methylcytidine (2'-C-MeC), a compound active against hepatitis C virus, was found to have activity against the 17D vaccine strain of YFV in cell culture (EC(90)=0.32 microg/ml, SI=141). This compound was effective when added as late as 16 h after virus challenge of Vero cells. When administered to YFV-infected hamsters 4 h prior to virus challenge at a dose as low as 80 mg/kg/d, 2'-C-MeC was effective in significantly improving survival and other disease parameters (weight change, serum ALT, and liver virus titers). Disease was improved when compound was administered beginning as late as 3 d post-virus infection. Broadly active antiviral compounds, such as 2'-C-MeC, represent potential for the development of compounds active against related viruses for the treatment of YFV.


Subject(s)
Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Cytidine/analogs & derivatives , Yellow Fever/drug therapy , Yellow fever virus/drug effects , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Body Weight , Chlorocebus aethiops , Cricetinae , Cytidine/administration & dosage , Cytidine/pharmacology , Female , Liver/virology , Mesocricetus , Survival Analysis , Treatment Outcome , Vero Cells , Viral Load
19.
Antiviral Res ; 86(2): 121-7, 2010 May.
Article in English | MEDLINE | ID: mdl-19874853

ABSTRACT

Several studies have reported favipiravir (T-705) to be effective in treating a number of viral diseases modeled in rodent systems. Notably, the related pyrazine derivative, T-1106, was found to be more effective than T-705 in treating yellow fever virus infection in hamsters. Based on these findings, we hypothesized that T-1106 may be more effective in treating hepatotropic Punta Toro virus (PTV, Phlebovirus) infection in rodents. In cell culture, the inhibitory concentrations of the compounds against various phleboviruses ranged from 3 to 55microM for T-705 and from 76 to 743microM for T-1106. In PTV-challenged hamsters, a model that generally presents with high liver viral loads, T-1106 was more effective at reducing mortality. However, in mice infected with PTV, a model wherein systemic infection is more prominent, the greater efficacy exhibited by T-1106 in the hamster system was not apparent. In contrast, T-705 was superior in preventing mortality in hamsters challenged with Pichinde virus (PICV, Arenavirus), an infection characterized as diffuse and pantropic. Remarkably, T-1106 has proven more active in vivo than would have been expected from our cell culture results, and our in vivo findings suggest that it is more effective in infections characterized predominantly by high levels of hepatic viral burden.


Subject(s)
Amides/therapeutic use , Antiviral Agents/therapeutic use , Bunyaviridae Infections/drug therapy , Nucleosides/therapeutic use , Pyrazines/therapeutic use , Animals , Chlorocebus aethiops , Cricetinae , Disease Models, Animal , Female , Liver/virology , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Serum/virology , Survival Analysis , Treatment Outcome , Vero Cells , Viral Load
20.
Antiviral Res ; 81(1): 37-46, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18840471

ABSTRACT

Cationic liposome-DNA complexes (CLDC) are cationic/neutral lipid carriers complexed with plasmid DNA that when administered systemically results in a robust T(H)1 cytokine response. CLDC have been shown to be effective in prophylaxis and therapeutic treatment of animal models of viral disease. To determine the contribution of liposomal delivery and CpG content of the plasmid DNA to the efficacy of CLDC; plasmid, CpG-free plasmid DNA, or CpG-containing oligodeoxynucleotides (ODN) with and without liposomes, as well as poly(I:C(12)U), were evaluated for their ability to elicit protection against lethal Punta Toro virus (PTV, Bunyaviridae, phlebovirus) challenge in hamsters. CLDC-containing plasmid significantly improved survival, decreased systemic and liver viral loads, and reduced liver damage due to progression of viral infection. Mouse-reactive ODNs complexed with liposomes failed to protect hamsters, whereas ODNs known to cross-react with human and mouse (CpG 2006) or non-liposomal poly(I:C(12)U) showed survival benefit but did not limit liver injury. Liposomes complexed with a non-CpG motif-containing plasmid reduced liver viral load and tissue damage, but did not protect hamsters from death. To evaluate the mechanisms of the enhanced activity of CLDC, microarray experiments examined differences in the gene expression profile. The results suggest a broad T(H)1 response elicited by liposomal delivery of a diverse sequence containing CpG and non-CpG elements may be a more effective antiviral treatment than other nucleic acid based immunotherapeutics.


Subject(s)
Oligodeoxyribonucleotides/administration & dosage , Phlebotomus Fever/immunology , Phlebotomus Fever/prevention & control , Phlebovirus/immunology , Animals , Cricetinae , Cytokines/blood , DNA/administration & dosage , DNA/chemistry , Female , Humans , Liposomes/chemistry , Liver/immunology , Liver/virology , Mesocricetus , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/chemistry , Phlebotomus Fever/therapy , Phlebotomus Fever/virology , Phlebovirus/genetics , Plasmids/administration & dosage , Plasmids/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...