Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Virol ; 98(4): e0011224, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38506509

RESUMEN

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.


Asunto(s)
Fiebre Hemorrágica Americana , Virus Junin , Vacunas Virales , Animales , Cobayas , Humanos , Ratones , Glicoproteínas/genética , Fiebre Hemorrágica Americana/prevención & control , Virus Junin/fisiología , Pueblos Sudamericanos , Vacunas Atenuadas/genética , Vacunas Virales/genética , Virulencia
2.
J Virol ; 98(4): e0013224, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38511932

RESUMEN

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.


Asunto(s)
Infecciones por Bunyaviridae , Bunyaviridae , Nucleótidos de Uracilo , Animales , Humanos , Ratones , Infecciones por Bunyaviridae/tratamiento farmacológico , Garrapatas , Estados Unidos , Nucleótidos de Uracilo/uso terapéutico
3.
Artículo en Inglés | MEDLINE | ID: mdl-33558299

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Guanosina Monofosfato/análogos & derivados , Guanosina/farmacología , Fosforamidas/farmacología , Profármacos/farmacología , SARS-CoV-2/efectos de los fármacos , Administración Oral , Animales , COVID-19/virología , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Coronavirus Humano 229E/metabolismo , Coronavirus Humano OC43/metabolismo , Cricetinae , Células Epiteliales/virología , Guanosina Monofosfato/farmacología , Humanos , Pulmón/virología , SARS-CoV-2/metabolismo , Células Vero , Replicación Viral/efectos de los fármacos
4.
PLoS Pathog ; 14(12): e1007439, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30576397

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Fiebre de Lassa , Virus Lassa/genética , Virulencia/efectos de los fármacos , Virulencia/genética , Animales , Chlorocebus aethiops , Farmacorresistencia Viral/efectos de los fármacos , Farmacorresistencia Viral/genética , Células HEK293 , Humanos , Virus Lassa/efectos de los fármacos , Ratones , Mutación , Células Vero , Proteínas del Envoltorio Viral/genética
5.
J Infect Dis ; 218(4): 522-527, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-29762684

RESUMEN

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.


Asunto(s)
Amidas/administración & dosificación , Antivirales/administración & dosificación , Coriomeningitis Linfocítica/tratamiento farmacológico , Virus de la Coriomeningitis Linfocítica/aislamiento & purificación , Pirazinas/administración & dosificación , Carga Viral , Animales , Modelos Animales de Enfermedad , Femenino , Huésped Inmunocomprometido , Coriomeningitis Linfocítica/virología , Masculino , Ratones Endogámicos NZB , Ribavirina/administración & dosificación , Análisis de Supervivencia , Receptores de Trasplantes , Resultado del Tratamiento
6.
J Virol ; 91(3)2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27881648

RESUMEN

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.


Asunto(s)
Infecciones por Bunyaviridae/virología , Modelos Biológicos , Phlebovirus/fisiología , Amidas/farmacología , Animales , Animales Modificados Genéticamente , Antivirales/farmacología , Infecciones por Bunyaviridae/tratamiento farmacológico , Infecciones por Bunyaviridae/genética , Infecciones por Bunyaviridae/mortalidad , Cricetinae , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Genotipo , Humanos , Fenotipo , Pirazinas/farmacología , Factor de Transcripción STAT2/genética
7.
Antimicrob Agents Chemother ; 56(8): 4168-74, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22615273

RESUMEN

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.


Asunto(s)
Adenoviridae/genética , Infecciones por Bunyaviridae/prevención & control , Interferón-alfa/genética , Interferón-alfa/metabolismo , Phlebovirus , Administración Intranasal , Animales , Antivirales/sangre , Antivirales/metabolismo , Cricetinae , Femenino , Vectores Genéticos , Interferón-alfa/sangre , Hígado/virología , Mesocricetus , Proteínas Recombinantes
8.
Cell Cycle ; 19(24): 3632-3638, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33305659

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Receptores Androgénicos/metabolismo , Receptores de Glucocorticoides/metabolismo , SARS-CoV-2/efectos de los fármacos , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/virología , Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/uso terapéutico , Línea Celular , Progresión de la Enfermedad , Regulación hacia Abajo , Glucocorticoides/antagonistas & inhibidores , Glucocorticoides/metabolismo , Humanos , Hidrocortisona/antagonistas & inhibidores , Factores Inmunológicos/farmacología , Factores Inmunológicos/uso terapéutico , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Receptores de Glucocorticoides/agonistas , Serina Endopeptidasas/metabolismo
9.
Antiviral Res ; 77(3): 215-24, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18222548

RESUMEN

Punta Toro virus (PTV) is a member of the Bunyaviridae family, genus Phlebovirus, related to the highly pathogenic Rift Valley fever virus (RVFV). It produces a disease in hamsters that models severe Rift Valley fever (RVF) in humans. The recent outbreak of RVF in Kenya stresses the need to identify prophylactic and therapeutic measures for preventing and treating severe forms of disease. To this end, interferon (IFN) alfacon-1 (consensus IFN-alpha) was evaluated in cell culture against RVFV and PTV, and in the hamster PTV infection model. Survival outcome following treatment initiated pre- and post-virus challenge and the suppression of viral burden and liver disease in infected hamsters was determined. Pre-treatment of cell cultures with IFN alfacon-1 induced marked antiviral activity against both viruses. Intraperitoneal treatment of hamsters initiated 4 h prior to infection with PTV was highly protective and greatly limited liver disease and systemic and liver viral burden. Complete protection from a highly lethal challenge dose was afforded by treatment initiated 36 h following viral inoculation. Although efficacy was much reduced, IFN alfacon-1 therapy was still beneficial when started as late as 3-5 days post-virus exposure. These studies suggest that IFN alfacon-1 may be an effective treatment for early intervention following infection with RVFV.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Bunyaviridae/tratamiento farmacológico , Infecciones por Bunyaviridae/prevención & control , Interferón Tipo I/uso terapéutico , Phlebovirus/efectos de los fármacos , Alanina Transaminasa/sangre , Animales , Antivirales/farmacología , Infecciones por Bunyaviridae/inmunología , Línea Celular , Supervivencia Celular , Cricetinae , Femenino , Haplorrinos , Interferón Tipo I/farmacología , Interferón-alfa , Ictericia/virología , Hígado/virología , Mesocricetus , Proteínas Recombinantes , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Suero/virología , Análisis de Supervivencia , Resultado del Tratamiento
10.
Antiviral Res ; 79(2): 105-13, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18423639

RESUMEN

Phenothiazine and derivatives were tested for inhibition of SARS-CoV replication. Phenothiazine slightly inhibited SARS-CoV replication in a neutral red (NR) uptake assay. Adding a propylamino group to give promazine reduced virus yields (VYR assay) with an EC(90)=8.3+/-2.8 microM, but without selectivity. Various substitutions in the basic phenothiazine structure did not promote efficacy. Phenazine ethosulfate was the most potent compound by VYR assay (EC(90)=6.1+/-4.3 microM). All compounds were toxic (IC(50)=6.6-74.5 microM) except for phenoxathiin (IC(50)=858+/-208 microM) and 10-(alpha-diethylamino-propionyl) phenothiazine.HCl (IC(50)=195+/-71.2 microM). Consequently, none were selective inhibitors of SARS-CoV replication (SI values <1-3.3 microM). These data portended the poor efficacy of promazine in a SARS-CoV mouse lung replication model. Intraperitoneal treatment with promazine using a prophylactic (-4h)/therapeutic regimen of 1, 10, or 50mg/(kg day) did not reduce virus lung titers at day 3, yet prolonged virus replication to 14 days. Similar therapeutic promazine doses were not efficacious. Thus, promazine did not affect SARS-CoV replication in vitro or in vivo, nor were any other phenothiazines efficacious in reducing virus replication. Therefore, treating SARS infections with compounds like promazine is not warranted.


Asunto(s)
Antivirales/farmacología , Fenotiazinas/farmacología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Replicación Viral , Animales , Antivirales/administración & dosificación , Antivirales/uso terapéutico , Antivirales/toxicidad , Supervivencia Celular , Quimioprevención , Chlorocebus aethiops , Citocinas/análisis , Femenino , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Rojo Neutro/metabolismo , Fenotiazinas/administración & dosificación , Fenotiazinas/uso terapéutico , Fenotiazinas/toxicidad , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/prevención & control , Células Vero
11.
Int Immunopharmacol ; 8(8): 1089-94, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18550012

RESUMEN

Recombinant Eimeria antigen (rEA) has been shown to have potent anticancer and antiviral activity in respective mouse disease models, presumably through robust immune stimulation that occurs via TLR11, a pattern recognition receptor that recognizes profilin-like proteins expressed on apicomplexan protozoans. Comparable immunostimulatory activity in other species has yet to be demonstrated. Since rEA is known to be highly effective in treating Punta Toro virus (PTV) infection in mice, its ability to elicit protective immunity in the hamster PTV infection model was investigated. rEA was given alone, or in combination with IL-18 or IL-2, and virally challenged hamsters were observed for mortality. Cytokine transcript profiles for IL-12p40, IL-21, IFN-gamma and TNF-alpha were assessed to evaluate the induction of these inflammatory mediators known to be induced in mice following exposure to rEA. A dose of 100 microg of rEA, given once 4 h prior to viral challenge, and a second time on day 3 of the infection, was found to be the most effective prophylactic therapy protecting 60% of treated hamsters from mortality, compared to only 5-10% observed in animals receiving placebo. Increased expression of IFN-gamma and IL-12p40 was evident following treatment with rEA. The data suggest that rEA does induce host antiviral responses in hamsters that result in significant protection from death, although determining the most appropriate dose for intervention in other species, including humans, will likely be challenging.


Asunto(s)
Antígenos de Protozoos/inmunología , Infecciones por Bunyaviridae/inmunología , Infecciones por Bunyaviridae/prevención & control , Eimeria/inmunología , Phlebovirus/inmunología , Animales , Antígenos de Protozoos/administración & dosificación , Infecciones por Bunyaviridae/mortalidad , Cricetinae , Femenino , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Subunidad p40 de la Interleucina-12/genética , Interleucina-18/administración & dosificación , Interleucina-18/inmunología , Interleucina-2/administración & dosificación , Interleucina-2/inmunología , Mesocricetus , Prohibitinas , Proteínas Recombinantes/inmunología , Transcripción Genética
12.
Antiviral Res ; 160: 48-54, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30339848

RESUMEN

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.


Asunto(s)
Antivirales/administración & dosificación , Antivirales/farmacología , Infecciones por Bunyaviridae/tratamiento farmacológico , Virus ADN/efectos de los fármacos , Desoxicitidina/análogos & derivados , Virus ARN/efectos de los fármacos , Estructuras Animales/virología , Animales , Peso Corporal , Efecto Citopatogénico Viral , Virus ADN/crecimiento & desarrollo , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Modelos Animales de Enfermedad , Ratones , Pruebas de Sensibilidad Microbiana , Placebos/administración & dosificación , Virus ARN/crecimiento & desarrollo , Análisis de Supervivencia , Resultado del Tratamiento , Carga Viral
13.
Antiviral Res ; 156: 38-45, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29864447

RESUMEN

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.


Asunto(s)
Antivirales/administración & dosificación , Nucleósidos de Purina/administración & dosificación , Fiebre del Valle del Rift/tratamiento farmacológico , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Adenina/análogos & derivados , Adenosina/análogos & derivados , Animales , Modelos Animales de Enfermedad , Inyecciones Intramusculares , Inyecciones Intraperitoneales , Hígado/virología , Mesocricetus , Pirrolidinas , Bazo/virología , Análisis de Supervivencia , Resultado del Tratamiento
14.
J Virol Methods ; 246: 51-57, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28359770

RESUMEN

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).


Asunto(s)
Antivirales/farmacología , Supervivencia Celular/efectos de los fármacos , Colorantes , Efecto Citopatogénico Viral , Virus ARN/efectos de los fármacos , Virus/efectos de los fármacos , Animales , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/patogenicidad , Virus Chikungunya/fisiología , Chlorocebus aethiops , Colorantes/química , Virus del Dengue/efectos de los fármacos , Virus del Dengue/patogenicidad , Virus del Dengue/fisiología , Virus Junin/efectos de los fármacos , Virus Junin/patogenicidad , Virus Junin/fisiología , Oxazinas , Virus ARN/patogenicidad , Virus ARN/fisiología , Células Vero , Replicación Viral/efectos de los fármacos , Xantenos
15.
Virology ; 511: 175-183, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28865344

RESUMEN

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.


Asunto(s)
Amidas/uso terapéutico , Antivirales/uso terapéutico , Infecciones por Bunyaviridae/patología , Infecciones por Bunyaviridae/prevención & control , Pirazinas/uso terapéutico , Factor de Transcripción STAT2/deficiencia , Estructuras Animales/patología , Animales , Quimioprevención , Cricetinae , Modelos Animales de Enfermedad , Inflamación/patología , Interferón Tipo I/inmunología , Resultado del Tratamiento
16.
Antiviral Res ; 69(3): 165-72, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16430975

RESUMEN

Cationic liposome-DNA complexes (CLDC) have been demonstrated to induce potent antitumor activities. The ability of these complexes to elicit protective immunity against viral infections has not been fully explored. Here we report findings on the use of CLDC as an antiviral agent in a mouse model of acute phleboviral (Punta Toro virus) disease. CLDC treatment of mice challenged with Punta Toro virus (PTV) resulted in dramatic increases in survival and reduced viral burden and other parameters indicative of protection against disease. CLDC were effective when administered by intraperitoneal and intravenous routes and elicited protective immunity when given within 1 day of virus challenge. Treatments administered 36 h or longer after challenge, however, were not effective in preventing mortality or disease. CLDC treatment induced release of a number of potential antiviral cytokines including IFN-gamma, IL-12, and IFN-alpha. Taken together, our findings indicate that non-specific immunotherapy with CLDC appears to be an effective treatment for blocking PTV-induced disease and suggests that further exploration in other viral disease models may be warranted.


Asunto(s)
Adyuvantes Inmunológicos , Infecciones por Bunyaviridae/prevención & control , Infecciones por Bunyaviridae/terapia , ADN Viral/inmunología , Liposomas/inmunología , Phlebovirus/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Infecciones por Bunyaviridae/inmunología , Infecciones por Bunyaviridae/virología , Citocinas/sangre , ADN Viral/administración & dosificación , Femenino , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Liposomas/administración & dosificación , Hígado/virología , Ratones , Análisis de Supervivencia , Carga Viral
17.
Antivir Chem Chemother ; 17(4): 175-83, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17066896

RESUMEN

Several arenaviruses endemic to South America (Junin, Machupo, and Guanarito) and Africa (Lassa) are known to cause frequently fatal haemorrhagic fever. With the exception of ribavirin, which has demonstrated efficacy in cases of Lassa fever, there is no other effective therapeutic for the treatment of arenaviral haemorrhagic fever. We have recently reported that consensus interferon-a (IFN alfacon-1) can protect hamsters from lethal Pichinde virus (PCV) infection, which serves as a model for acute arenaviral disease in humans. Here we demonstrate highly effective therapy through the combined use of ribavirin with IFN alfacon-1 for the treatment of PCV infection in hamsters. Ribavirin was given orally, twice per day for 7 days, and IFN alfacon-1 was administered intraperitoneally once per day for 10 days. Treatments were initiated 1-5 days post-virus challenge using various dose combinations, many of which were less than optimal when the drugs were given independently. Combining suboptimal doses of ribavirin (5-10 mg/kg/day) with IFN alfacon-1 (5-10 microg/kg/day), we were able to demonstrate increased protection from mortality, reduced viral burden and liver disease, and greatly extended survival times as compared to treatments where drugs were administered alone. Our data indicate that combination therapy results in synergistic activity that may slow down the progression of the disease and decrease fatality rates associated with severe arenaviral infections in humans. Further, combination therapy reduces the effective dosage of ribavirin, which would serve to limit its toxicity.


Asunto(s)
Infecciones por Arenaviridae/tratamiento farmacológico , Interferón Tipo I/uso terapéutico , Ribavirina/uso terapéutico , Enfermedad Aguda/mortalidad , Administración Oral , Animales , Antivirales/uso terapéutico , Infecciones por Arenaviridae/mortalidad , Cricetinae , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Femenino , Interferón Tipo I/administración & dosificación , Interferón-alfa , Placebos , Proteínas Recombinantes , Ribavirina/administración & dosificación , Análisis de Supervivencia , Factores de Tiempo
18.
Antiviral Res ; 126: 62-8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26711718

RESUMEN

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.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Fiebres Hemorrágicas Virales/tratamiento farmacológico , Pirazinas/farmacología , Virus ARN/efectos de los fármacos , Ribavirina/farmacología , Animales , Arenavirus/efectos de los fármacos , Chlorocebus aethiops , Cricetinae , Virus del Dengue/efectos de los fármacos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Cobayas , Orthohantavirus/efectos de los fármacos , Virus de la Fiebre Hemorrágica de Crimea-Congo/efectos de los fármacos , Fiebre Hemorrágica Americana/tratamiento farmacológico , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebres Hemorrágicas Virales/sangre , Fiebres Hemorrágicas Virales/veterinaria , Fiebres Hemorrágicas Virales/virología , Virus Junin/efectos de los fármacos , Masculino , Mesocricetus , Ratones , Células Vero
19.
Front Microbiol ; 6: 651, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26175722

RESUMEN

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.

20.
Antivir Chem Chemother ; 23(4): 151-9, 2014 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23337126

RESUMEN

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.


Asunto(s)
Antivirales/química , Antivirales/uso terapéutico , Arenavirus del Nuevo Mundo/efectos de los fármacos , Fiebre Hemorrágica Americana/tratamiento farmacológico , Animales , Chlorocebus aethiops , Humanos , Virus Junin/efectos de los fármacos , Ratones , Fenoles/química , Fenoles/uso terapéutico , Sulfuros/química , Sulfuros/uso terapéutico , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA