Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
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 ; 95(14): e0039721, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-33952638

RESUMEN

Live-attenuated virus vaccines are highly effective in preventing viral disease but carry intrinsic risks of residual virulence and reversion to pathogenicity. The classically derived Candid#1 virus protects seasonal field workers in Argentina against zoonotic infection by Junín virus (JUNV) but is not approved in the United States, in part due to the potential for reversion at the attenuating locus, a phenylalanine-to-isoleucine substitution at position 427 in the GP2 subunit of the GPC envelope glycoprotein. Previously, we demonstrated facile reversion of recombinant Candid#1 (rCan) in cell culture and identified an epistatic interaction between the attenuating I427 and a secondary K33S mutation in the stable signal peptide (SSP) subunit of GPC that imposes an evolutionary barrier to reversion. The magnitude of this genetic barrier is manifest in our repeated failures to rescue the hypothetical revertant virus. In this study, we show that K33S rCan is safe and attenuated in guinea pigs and capable of eliciting potent virus-neutralizing antibodies. Immunized animals are fully protected against lethal challenge with virulent JUNV. In addition, we employed a more permissive model of infection in neonatal mice to investigate genetic reversion. RNA sequence analysis of the recovered virus identified revertant viruses in pups inoculated with the parental rCan virus and none in mice receiving K33S rCan (P < 0.0001). Taken together, our findings support the further development of K33S rCan as a safe second-generation JUNV vaccine. IMPORTANCE Our most successful vaccines comprise weakened strains of virus that initiate a limited and benign infection in immunized persons. The live-attenuated Candid#1 strain of Junín virus (JUNV) was developed to protect field workers in Argentina from rodent-borne hemorrhagic fever but is not licensed in the United States, in part due to the likelihood of genetic reversion to virulence. A single-amino-acid change in the GPC envelope glycoprotein of the virus is responsible for attenuation, and a single nucleotide change may regenerate the pathogenic virus. Here, we take advantage of a unique genetic interaction between GPC subunits to design a mutant Candid#1 virus that establishes an evolutionary barrier to reversion. The mutant virus (K33S rCan) is fully attenuated and protects immunized guinea pigs against lethal JUNV infection. We find no instances of reversion in mice inoculated with K33S rCan. This work supports the further development of K33S rCan as a second-generation JUNV vaccine.


Asunto(s)
Fiebre Hemorrágica Americana/prevención & control , Virus Junin/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Chlorocebus aethiops , Cobayas , Fiebre Hemorrágica Americana/inmunología , Inmunogenicidad Vacunal , Virus Junin/genética , Virus Junin/patogenicidad , Masculino , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Células Vero , Vacunas Virales/genética , Virulencia
3.
Antiviral Res ; : 105952, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38945484

RESUMEN

Argentine hemorrhagic fever, caused by Junín virus (JUNV), is the most common of the South American arenaviral hemorrhagic fevers. The disease has a case fatality rate of 15-30% in untreated patients. Although early intervention with immune plasma is effective, diminishing stocks and limited availability outside of Argentina underscores the need for new therapeutics. Ideally, these would be broadly active agents effective against all the pathogenic arenaviruses. The fusion inhibitor LHF-535 and the nucleoside analog favipiravir have shown promise in animal models of Lassa fever, a disease endemic in parts of Africa and the most prominent of the arenaviral hemorrhagic fevers. Against JUNV, a high dose of favipiravir is required to achieve protection in the gold-standard guinea pig infection model. Here, we demonstrate a synergistic effect by the coadministration of LHF-535 with a sub-optimal dose of favipiravir in guinea pigs challenged with JUNV. Administered individually, LHF-535 and sub-optimal favipiravir only delayed the onset of severe disease. However, combined dosing of the drugs afforded complete protection against lethal JUNV infection in guinea pigs. The benefits of the drug combination were also evident by the absence of viremia and infectious virus in tissues compared to guinea pigs treated with only the placebos. Thus, combined targeting of JUNV-endosomal membrane fusion and the viral polymerase with pan-arenaviral LHF-535 and favipiravir may expand their indication beyond Lassa fever, providing a significant barrier to drug resistance.

4.
Antiviral Res ; 208: 105444, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36243175

RESUMEN

Infections by pathogenic New World mammarenaviruses (NWM)s, including Junín virus (JUNV), can result in a severe life-threatening viral hemorrhagic fever syndrome. In the absence of FDA-licensed vaccines or antivirals, these viruses are considered high priority pathogens. The mammarenavirus envelope glycoprotein complex (GPC) mediates pH-dependent fusion between viral and cellular membranes, which is essential to viral entry and may be vulnerable to small-molecule inhibitors that disrupt this process. ARN-75039 is a potent fusion inhibitor of a broad spectrum of pseudotyped and native mammarenaviruses in cell culture and Tacaribe virus infection in mice. In the present study, we evaluated ARN-75039 against pathogenic JUNV in the rigorous guinea pig infection model. The compound was well-tolerated and had favorable pharmacokinetics supporting once-per-day oral dosing in guinea pigs. Importantly, significant protection against JUNV challenge was observed even when ARN-75039 was withheld until 6 days after the viral challenge when clinical signs of disease are starting to develop. We also show that ARN-75039 combination treatment with favipiravir, a viral polymerase inhibitor, results in synergistic activity in vitro and improves survival outcomes in JUNV-challenged guinea pigs. Our findings support the continued development of ARN-75039 as an attractive therapeutic candidate for treating mammarenaviral hemorrhagic fevers, including those associated with NWM infection.


Asunto(s)
Arenaviridae , Fiebre Hemorrágica Americana , Fiebres Hemorrágicas Virales , Virus Junin , Cobayas , Ratones , Animales , Fiebre Hemorrágica Americana/tratamiento farmacológico , Pirazinas/farmacología , Pirazinas/uso terapéutico , Amidas/farmacología , Amidas/uso terapéutico , Antirretrovirales/farmacología
5.
Nat Commun ; 13(1): 558, 2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35091550

RESUMEN

Five New World mammarenaviruses (NWMs) cause life-threatening hemorrhagic fever (HF). Cellular entry by these viruses is mediated by human transferrin receptor 1 (hTfR1). Here, we demonstrate that an antibody (ch128.1/IgG1) which binds the apical domain of hTfR1, potently inhibits infection of attenuated and pathogenic NWMs in vitro. Computational docking of the antibody Fab crystal structure onto the known structure of hTfR1 shows an overlapping receptor-binding region shared by the Fab and the viral envelope glycoprotein GP1 subunit that binds hTfR1, and we demonstrate competitive inhibition of NWM GP1 binding by ch128.1/IgG1 as the principal mechanism of action. Importantly, ch128.1/IgG1 protects hTfR1-expressing transgenic mice against lethal NWM challenge. Additionally, the antibody is well-tolerated and only partially reduces ferritin uptake. Our findings provide the basis for the development of a novel, host receptor-targeted antibody therapeutic broadly applicable to the treatment of HF of NWM etiology.


Asunto(s)
Antígenos CD/metabolismo , Arenaviridae/metabolismo , Fiebre Hemorrágica Americana/metabolismo , Receptores de Transferrina/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Células A549 , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Arenaviridae/efectos de los fármacos , Arenaviridae/fisiología , Chlorocebus aethiops , Fiebre Hemorrágica Americana/prevención & control , Fiebre Hemorrágica Americana/virología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Virus Junin/efectos de los fármacos , Virus Junin/fisiología , Ratones Endogámicos C57BL , Ratones Transgénicos , Simulación del Acoplamiento Molecular , Unión Proteica/efectos de los fármacos , Receptores de Transferrina/antagonistas & inhibidores , Receptores de Transferrina/inmunología , Células Vero
6.
Am J Trop Med Hyg ; 107(5): 1091-1098, 2022 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-36122681

RESUMEN

The Rift Valley fever virus (RVFV) MP-12 vaccine is a promising human and veterinary vaccine. Although the vaccine elicited neutralizing antibody (nAb) in human volunteers, the minimal antibody titer that is needed to afford protection is unknown. Therefore, this study was conducted to determine the minimal nAb titer elicited by the RVFV MP-12 vaccine in human volunteers that protected mice against lethal RVFV challenge as a surrogate assessment of the protective efficacy of the vaccine. Among volunteers who were vaccinated with the MP-12 vaccine during a phase II trial, sera with antibody titers of 1:20 collected 5 years post-vaccination (PV), 1:40 titer collected 2 years PV, and 1:80 titer collected 1 year PV was passively transferred to groups of BALB/c mice. Blood samples were obtained 1 day after passive transfer to determine the RVFV neutralizing nAb titer before challenge with pathogenic RVFV (strain ZH501). Our results indicated that 1 day after passive transfer of the immune sera, an approximate 4-fold reduction in circulating nAb titers was detected in the mice. The presence of RVFV nAb titers in the range of 1:5 to 1:20 were generally protective (75-100% survival). These results suggested that circulating titers of 1:5 or higher offer a high degree of protection by MP-12-elicited antibody in human volunteers. Also, the findings highlighted the value of using the BALB/c mouse RVFV challenge model as a surrogate for evaluating the protective nAb responses elicited by MP-12 and possible use for evaluating the efficacy of other RVFV vaccine candidates.


Asunto(s)
Fiebre del Valle del Rift , Virus de la Fiebre del Valle del Rift , Vacunas Virales , Ratones , Humanos , Animales , Voluntarios Sanos , Vacunas Atenuadas , Anticuerpos Antivirales , Anticuerpos Neutralizantes , Ratones Endogámicos BALB C , Modelos Animales de Enfermedad
7.
Antimicrob Agents Chemother ; 54(1): 126-33, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19901093

RESUMEN

Favipiravir (T-705 [6-fluoro-3-hydroxy-2-pyrazinecarboxamide]) and oseltamivir were combined to treat influenza virus A/NWS/33 (H1N1), A/Victoria/3/75 (H3N2), and A/Duck/MN/1525/81 (H5N1) infections. T-705 alone inhibited viruses in cell culture at 1.4 to 4.3 microM. Oseltamivir inhibited these three viruses in cells at 3.7, 0.02, and 0.16 microM and in neuraminidase assays at 0.94, 0.46, and 2.31 nM, respectively. Oral treatments were given twice daily to mice for 5 to 7 days starting, generally, 24 h after infection. Survival resulting from 5 days of oseltamivir treatment (0.1 and 0.3 mg/kg/day) was significantly better in combination with 20 mg/kg of body weight/day of T-705 against the H1N1 infection. Treatment of the H3N2 infection required 50 mg/kg/day of oseltamivir for 7 days to achieve 60% protection; 25 mg/kg/day was ineffective. T-705 was >or=70% protective at 50 to 100 mg/kg/day but inactive at 25 mg/kg/day. The combination of inhibitors (25 mg/kg/day each) increased survival to 90%. The H5N1 infection was not benefited by treatment with oseltamivir (

Asunto(s)
Amidas/uso terapéutico , Antivirales/uso terapéutico , Virus de la Influenza A , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Oseltamivir/uso terapéutico , Pirazinas/uso terapéutico , Animales , Línea Celular , Combinación de Medicamentos , Interacciones Farmacológicas , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/enzimología , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/enzimología , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/enzimología , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/enzimología , Ratones , Ratones Endogámicos BALB C , Neuraminidasa/antagonistas & inhibidores , Infecciones por Orthomyxoviridae/virología
8.
Elife ; 92020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32452770

RESUMEN

Junín virus (JUNV) is one of five New World mammarenaviruses (NWMs) that causes fatal hemorrhagic disease in humans and is the etiological agent of Argentine hemorrhagic fever (AHF). The pathogenesis underlying AHF is poorly understood; however, a prolonged, elevated interferon-α (IFN-α) response is associated with a negative disease outcome. A feature of all NWMs that cause viral hemorrhagic fever is the use of human transferrin receptor 1 (hTfR1) for cellular entry. Here, we show that mice expressing hTfR1 develop a lethal disease course marked by an increase in serum IFN-α concentration when challenged with JUNV. Further, we provide evidence that the type I IFN response is central to the development of severe JUNV disease in hTfR1 mice. Our findings identify hTfR1-mediated entry and the type I IFN response as key factors in the pathogenesis of JUNV infection in mice.


Asunto(s)
Antígenos CD/fisiología , Fiebre Hemorrágica Americana/virología , Interacciones Huésped-Patógeno , Interferón-alfa/fisiología , Virus Junin/fisiología , Receptores de Transferrina/fisiología , Animales , Ratones
9.
Antimicrob Agents Chemother ; 53(5): 2120-8, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19273672

RESUMEN

An amantadine-resistant influenza A/Duck/MN/1525/81 (H5N1) virus was developed from the low-pathogenic North American wild-type (amantadine-sensitive) virus for studying treatment of infections in cell culture and in mice. Double combinations of amantadine, oseltamivir (or the cell culture-active form, oseltamivir carboxylate), and ribavirin were used. Amantadine-oseltamivir carboxylate and amantadine-ribavirin combinations showed synergistic interactions over a range of doses against wild-type virus in Madin-Darby canine kidney (MDCK) cell culture, but oseltamivir carboxylate-ribavirin combinations did not. Primarily additive interactions were seen with oseltamivir carboxylate-ribavirin combinations against amantadine-resistant virus. The presence of amantadine in drug combinations against the resistant virus did not improve activity. The wild-type and amantadine-resistant viruses were lethal to mice by intranasal instillation. The resistant virus infection could not be treated with amantadine up to 100 mg/kg body weight/day, whereas the wild-type virus infection was treatable with oral doses of 10 (weakly effective) to 100 mg/kg/day administered twice a day for 5 days starting 4 h prior to virus exposure. Drug combination studies showed that treatment of the amantadine-resistant virus infection with amantadine-oseltamivir or amantadine-ribavirin combinations was not significantly better than using oseltamivir or ribavirin alone. In contrast, the oseltamivir-ribavirin (25- and 75-mg/kg/day combination) treatments produced significant reductions in mortality. The wild-type virus infection was markedly reduced in severity by all three combinations (amantadine, 10 mg/kg/day combined with the other compounds at 20 or 40 mg/kg/day) compared to monotherapy with the three compounds. Results indicate a lack of benefit of amantadine in combinations against amantadine-resistant virus, but positive benefits in combinations against amantadine-sensitive virus.


Asunto(s)
Amantadina , Antivirales , Modelos Animales de Enfermedad , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Oseltamivir , Ribavirina , Amantadina/administración & dosificación , Amantadina/farmacología , Amantadina/uso terapéutico , Animales , Antivirales/administración & dosificación , Antivirales/farmacología , Antivirales/uso terapéutico , Línea Celular , Interacciones Farmacológicas , Farmacorresistencia Viral/genética , Quimioterapia Combinada , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana/métodos , Infecciones por Orthomyxoviridae/virología , Oseltamivir/administración & dosificación , Oseltamivir/farmacología , Oseltamivir/uso terapéutico , Ribavirina/administración & dosificación , Ribavirina/farmacología , Ribavirina/uso terapéutico , Organismos Libres de Patógenos Específicos , Resultado del Tratamiento
10.
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
11.
Antiviral Res ; 73(1): 69-77, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16712967

RESUMEN

A novel carbocyclic thymidine analog, N-methanocarbathymidine [(N)-MCT], was evaluated for inhibition of orthopoxvirus infections. Efficacy in vitro was assessed by plaque reduction assays against wild-type and cidofovir-resistant strains of cowpox and vaccinia viruses in nine different cell lines. Minimal differences were seen in antiviral activity against wild-type and cidofovir-resistant viruses. (N)-MCT's efficacy was affected by the cell line used for assay, with 50% poxvirus-inhibitory concentrations in cells as follows: mouse=0.6-2.2 microM, rabbit=52-90 microM, monkey=87 to >1000 microM, and human=39-220 microM. Limited studies performed with carbocyclic thymidine indicated a similar cell line dependency for antiviral activity. (N)-MCT did not inhibit actively dividing uninfected cells at 1000 microM. The potency of (N)-MCT against an S-variant thymidine kinase-deficient vaccinia virus was similar to that seen against S-variant and wild-type viruses in mouse, monkey, and human cells, implicating a cellular enzyme in the phosphorylation of the compound. Mice were intranasally infected with cowpox and vaccinia viruses followed 24h later by intraperitoneal treatment with (N)-MCT (twice a day for 7 days) or cidofovir (once a day for 2 days). (N)-MCT treatment at 100 and 30 mg/kg/day resulted in 90 and 20% survival from cowpox virus infection, respectively, compared to 0% survival in the placebo group. Statistically significant reductions in lung virus titers on day 5 occurred in 10, 30, and 100mg/kg/day treated mice. These same doses were also active against a lethal vaccinia virus (WR strain) challenge, and protection was seen down to 10mg/kg/day against a lethal vaccinia virus (IHD strain) infection. Cidofovir (100mg/kg/day) protected animals from death in all three infections.


Asunto(s)
Antivirales/uso terapéutico , Viruela Vacuna/tratamiento farmacológico , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Timidina/análogos & derivados , Vaccinia/tratamiento farmacológico , Animales , Antivirales/farmacología , Línea Celular , Viruela Vacuna/virología , Virus de la Viruela Vacuna/efectos de los fármacos , Virus de la Viruela Vacuna/crecimiento & desarrollo , Humanos , Ratones , Ratones Endogámicos BALB C , Conejos , Infecciones del Sistema Respiratorio/virología , Timidina/farmacología , Timidina/uso terapéutico , Vaccinia/virología , Virus Vaccinia/efectos de los fármacos , Virus Vaccinia/crecimiento & desarrollo
12.
Antiviral Res ; 145: 131-135, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28780425

RESUMEN

A collection of Old and New World arenaviruses are etiologic agents of viral hemorrhagic fever, a syndrome that features hematologic abnormalities, vascular leak, hypovolemia, and multi-organ failure. Treatment is limited to ribavirin for Lassa fever and immune plasma for Argentine hemorrhagic fever. Improved therapeutic options that are safe, more effective and widely available are needed. Here, we show that modification of favipiravir treatment to include a high-dose loading period achieves complete protection in a guinea pig model of Argentine hemorrhagic fever when treatment was initiated two days following challenge with Junin virus (JUNV). This loading dose strategy also protected 50% of animals from lethal disease when treatment was delayed until 5 days post-infection and extended the survival time in those that succumbed. Consistent with the survival data, dramatic reductions in serum and tissue virus loads were observed in animals treated with favipiravir. This is the first report demonstrating complete protection against uniformly lethal JUNV infection in guinea pigs by administration of a small molecule antiviral drug.


Asunto(s)
Amidas/administración & dosificación , Antivirales/administración & dosificación , Fiebre Hemorrágica Americana/tratamiento farmacológico , Virus Junin/efectos de los fármacos , Pirazinas/administración & dosificación , Amidas/uso terapéutico , Animales , Antivirales/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Cobayas , Fiebre Hemorrágica Americana/sangre , Fiebre Hemorrágica Americana/mortalidad , Pirazinas/uso terapéutico , Análisis de Supervivencia , Carga Viral/efectos de los fármacos
13.
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
14.
Antivir Chem Chemother ; 17(4): 185-92, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17066897

RESUMEN

Mouse models have been widely used for evaluating potential influenza virus inhibitors. However, the viral strains traditionally used in these models are fairly old and do not represent currently circulating viruses in nature. We developed two new lethal infection models in mice using mouse-adapted influenza A/New Caledonia/20/99 (H1N1) and influenza B/Sichuan/379/99 viruses. Both virus infections were used to study oral treatment with oseltamivir and ribavirin, both alone and in combination. Oral treatments were given twice daily for 5 days starting 4 h before infection in initial studies. Against influenza A, oseltamivir was active at 10, 20, and 40 mg/kg/day, protected 80-100% of mice from death and reduced lung consolidation - ribavirin was similarly effective at 20, 40, and 80 mg/kg/day. When treatments were initiated after virus challenge, delaying treatment with oseltamivir even 1 day caused it to be ineffective. Ribavirin prevented mortality by 50-80% when treatments were delayed 1-4 days after infection. The combination of the two drugs (oseltamivir at 20 mg/kg/day and ribavirin at 40 mg/kg/day) was no better than ribavirin alone. In contrast to what we observed with influenza A virus infections, oseltamivir and ribavirin showed similar dose-related antiviral activities against influenza B virus infections. The compounds both significantly increased survival when treatments started up to 4 days after infection, but ribavirin was more active than oseltamivir (50-80% survival compared to 30-40% survival, respectively, when starting treatments on days 2-4 after infection). By varying the doses of each drug that were used in combination (oseltamivir at 1.25, 2.5 and 5 mg/kg/day; ribavirin at 5, 10 and 20 mg/kg/day) certain dosage combinations were superior to either compound used alone as assessed by decreased mortality, lung virus titre, lung score and lung weight parameters. These activities differed from published results with older, more established virus strains as oseltamivir was less effective and ribavirin was more active than previously reported.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza B/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Oseltamivir/uso terapéutico , Ribavirina/uso terapéutico , Administración Oral , Animales , Antivirales/administración & dosificación , Antivirales/uso terapéutico , Células Cultivadas , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Sinergismo Farmacológico , Quimioterapia Combinada , Ratones , Oseltamivir/administración & dosificación , Placebos , Ribavirina/administración & dosificación , Factores de Tiempo
15.
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
16.
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
17.
Antivir Chem Chemother ; 16(3): 203-11, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16004083

RESUMEN

The wild-type (WT) vaccinia (WR strain) virus is highly virulent to mice by intranasal inoculation, yet death can be prevented by cidofovir treatment. A cidofovir-resistant (CDV-R) mutant of the virus was developed by 15 Vero cell culture passages in order to determine cross-resistance to other inhibitors, growth characteristics, virulence in infected mice, and suitability of the animal model for studying antiviral therapies. Comparisons were made to the original WT virus and to a WT virus passaged 15 times in culture (WTp15 virus). Cidofovir inhibited WT, WTp15, and CDV-R viruses by 50% at 61, 56 and 790 microM, respectively, in plaque reduction assays, with similar inhibition seen in virus yield studies. Cross-resistance occurred with compounds related to cidofovir, but not with unrelated nucleosides. The resistant virus produced 300-fold fewer infectious particles (PFU) than WT and WTp15 viruses in mouse C1271 cells, yet replicated similarly in Vero (monkey) cells. The CDV-R virus was completely attenuated for virulence at 10(7) PFU per mouse in normal BALB/c mice and in severe combined immunodeficient (SCID) mice. The WTp15 virus was 100-fold less virulent than WT virus in BALB/c mice. Thus, the lack of virulence of the resistant virus in the animal model is explained partly by its reduced ability to replicate in mouse cells and by attenuation occurring as a result of extensive cell culturing (inferred from what occurred with the WTp15 virus). Lung and snout virus titre reduction parameters were used to assess antiviral activity of compounds in BALB/c mice infected intranasally with the CDV-R virus. Cidofovir, HDP-cidofovir and arabinofuranosyladenine treatments reduced lung virus titres or = eight-fold. The animal model appears to have limited utility in drug efficacy testing.


Asunto(s)
Citosina/análogos & derivados , Modelos Animales de Enfermedad , Resistencia a Medicamentos , Organofosfonatos/farmacología , Vaccinia , Animales , Células Cultivadas , Cidofovir , Citosina/farmacología , Resistencia a Medicamentos/genética , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Mutación , Vaccinia/virología , Virus Vaccinia/efectos de los fármacos , Virus Vaccinia/genética , Virus Vaccinia/patogenicidad , Replicación Viral
18.
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.

19.
PLoS One ; 10(1): e0116722, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25607955

RESUMEN

Rift Valley fever virus (RVFV) is a formidable pathogen that causes severe disease and abortion in a variety of livestock species and a range of disease in humans that includes hemorrhagic fever, fulminant hepatitis, encephalitis and blindness. The natural transmission cycle involves mosquito vectors, but exposure can also occur through contact with infected fluids and tissues. The lack of approved antiviral therapies and vaccines for human use underlies the importance of small animal models for proof-of-concept efficacy studies. Several mouse and rat models of RVFV infection have been well characterized and provide useful systems for the study of certain aspects of pathogenesis, as well as antiviral drug and vaccine development. However, certain host-directed therapeutics may not act on mouse or rat pathways. Here, we describe the natural history of disease in golden Syrian hamsters challenged subcutaneously with the pathogenic ZH501 strain of RVFV. Peracute disease resulted in rapid lethality within 2 to 3 days of RVFV challenge. High titer viremia and substantial viral loads were observed in most tissues examined; however, histopathology and immunostaining for RVFV antigen were largely restricted to the liver. Acute hepatocellular necrosis associated with a strong presence of viral antigen in the hepatocytes indicates that fulminant hepatitis is the likely cause of mortality. Further studies to assess the susceptibility and disease progression following respiratory route exposure are warranted. The use of the hamsters to model RVFV infection is suitable for early stage antiviral drug and vaccine development studies.


Asunto(s)
Hígado/patología , Hígado/virología , Mesocricetus/virología , Fiebre del Valle del Rift/mortalidad , Virus de la Fiebre del Valle del Rift/patogenicidad , Animales , Antígenos Virales/metabolismo , Células Cultivadas , Femenino , Hepatitis Animal/mortalidad , Hepatitis Animal/patología , Hepatitis Animal/virología , Fiebre del Valle del Rift/patología , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/inmunología , Enfermedades de los Roedores/mortalidad , Enfermedades de los Roedores/patología , Enfermedades de los Roedores/virología , Carga Viral , Viremia/virología
20.
Antiviral Res ; 54(2): 113-20, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12062396

RESUMEN

The acyclic purine nucleoside analog, 2-amino-7-[(1,3-dihydroxy-2-propoxy)methyl]purine (S2242) and its orally active diacetate ester prodrug (HOE961) were reported to be potent inhibitors of vaccinia virus replication in cell culture and in infected mice. These compounds were evaluated further, using infections with the related cowpox virus. Against a wild-type (WT) cowpox virus strain in mouse C127I cell culture, 50% effective concentrations (EC(50), determined by plaque reduction assays) of S2242 and cidofovir (a positive control) were 3.5 and 1.0 microM, respectively. EC(50) values obtained against a cidofovir-resistant strain of the virus were 33 and 230 microM, respectively. Compounds were at least ten-fold less potent against WT virus in Vero cells than C127I cells. S2242 and cidofovir were 50% inhibitory to the proliferation of uninfected C127I cells at 340 and 180 microM, respectively, but neither compound inhibited Vero cell growth at 1000 microM. Mice were lethally infected with cowpox virus by intranasal inoculation, followed 24 h later by antiviral treatment for 5 consecutive days. Once or twice daily intraperitoneal (i.p.) treatments with either S2242 or HOE961 at 100 mg/kg per day resulted in > or = 70 survival compared with no survivors in the placebo group. Lower doses of these compounds (10 and 30 mg/kg per day) were not protective, however. Cidofovir was 100% protective at 30 mg/kg per day. A 10-day course of treatment gave comparable survival results and demonstrated the oral efficacy of HOE961. Treatments with S2242 (100 mg/kg per day) and cidofovir (30 mg/kg per day) each reduced lung and nasal virus titers by approximately ten-fold, whereas, HOE961 (100 mg/kg per day) was less active. Overall, S2242 and HOE961 were found to be effective against cowpox virus infections in mice but were less potent than cidofovir. Since, HOE961 was orally active, it may have advantages over the other parenterally administered compounds for treating orthopoxvirus infections.


Asunto(s)
Antivirales/farmacología , Virus de la Viruela Vacuna/crecimiento & desarrollo , Viruela Vacuna/tratamiento farmacológico , Organofosfonatos , Profármacos/farmacología , Purinas/farmacología , Animales , Área Bajo la Curva , Peso Corporal , Chlorocebus aethiops , Cidofovir , Virus de la Viruela Vacuna/metabolismo , Citosina/análogos & derivados , Citosina/farmacología , Modelos Animales de Enfermedad , Femenino , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Mucosa Nasal/virología , Compuestos Organofosforados/farmacología , Estadísticas no Paramétricas , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA