Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Virol J ; 10: 221, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23816343

RESUMO

BACKGROUND: Tacaribe virus (TCRV) is a less biohazardous relative of the highly pathogenic clade B New World arenaviruses that cause viral hemorrhagic fever syndromes and require handling in maximum containment facilities not readily available to most researchers. AG129 type I and II interferon receptor knockout mice have been shown to be susceptible to TCRV infection, but the pathogenic mechanisms contributing to the lethal disease are unclear. METHODS: To gain insights into the pathogenesis of TCRV infection in AG129 mice, we assessed hematologic and cytokine responses during the course of infection, as well as changes in the permeability of the vascular endothelium. We also treated TCRV-challenged mice with MY-24, a compound that prevents mortality without affecting viral loads during the acute infection, and measured serum and tissue viral titers out to 40 days post-infection to determine whether the virus is ultimately cleared in recovering mice. RESULTS: We found that the development of viremia and splenomegaly precedes an elevation in white blood cells and the detection of high levels of proinflammatory mediators known to destabilize the endothelial barrier, which likely contributes to the increased vascular permeability and weight loss that was observed several days prior to when the mice generally succumb to TCRV challenge. In surviving mice treated with MY-24, viremia and liver virus titers were not cleared until 2-3 weeks post-infection, after which the mice began to recover lost weight. Remarkably, substantial viral loads were still present in the lung, spleen, brain and kidney tissues at the conclusion of the study. CONCLUSIONS: Our findings suggest that vascular leak may be a contributing factor in the demise of TCRV-infected mice, as histopathologic findings are generally mild to moderate in nature, and as evidenced with MY-24 treatment, animals can survive in the face of high viral loads.


Assuntos
Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/patologia , Arenavirus do Novo Mundo/imunologia , Arenavirus do Novo Mundo/patogenicidade , Permeabilidade Capilar , Citocinas/metabolismo , Estruturas Animais/virologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Carga Viral , Viremia/imunologia , Viremia/patologia
2.
Antimicrob Agents Chemother ; 56(8): 4168-74, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22615273

RESUMO

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.


Assuntos
Adenoviridae/genética , Infecções por Bunyaviridae/prevenção & controle , Interferon-alfa/genética , Interferon-alfa/metabolismo , Phlebovirus , Administração Intranasal , Animais , Antivirais/sangue , Antivirais/metabolismo , Cricetinae , Feminino , Vetores Genéticos , Interferon-alfa/sangue , Fígado/virologia , Mesocricetus , Proteínas Recombinantes
3.
Antimicrob Agents Chemother ; 54(1): 126-33, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19901093

RESUMO

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 (

Assuntos
Amidas/uso terapêutico , Antivirais/uso terapêutico , Vírus da Influenza A , Infecções por Orthomyxoviridae/tratamento farmacológico , Oseltamivir/uso terapêutico , Pirazinas/uso terapêutico , Animais , Linhagem Celular , Combinação de Medicamentos , Interações Medicamentosas , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/enzimologia , Vírus da Influenza A Subtipo H3N2/efeitos dos fármacos , Vírus da Influenza A Subtipo H3N2/enzimologia , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/enzimologia , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/enzimologia , Camundongos , Camundongos Endogâmicos BALB C , Neuraminidase/antagonistas & inibidores , Infecções por Orthomyxoviridae/virologia
4.
Virol J ; 7: 240, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20846417

RESUMO

BACKGROUND: A number of RNA viruses cause viral hemorrhagic fever (VHF), in which proinflammatory mediators released from infected cells induce increased permeability of the endothelial lining of blood vessels, leading to loss of plasma volume, hypotension, multi-organ failure, shock and death. The optimal treatment of VHF should therefore include both the use of antiviral drugs to inhibit viral replication and measures to prevent or correct changes in vascular function. Although rodent models have been used to evaluate treatments for increased vascular permeability (VP) in bacterial sepsis, such studies have not been performed for VHF. RESULTS: Here, we use an established model of Pichinde virus infection of hamsters to demonstrate how changes in VP can be detected by intravenous infusion of Evans blue dye (EBD), and compare those measurements to changes in hematocrit, serum albumin concentration and serum levels of proinflammatory mediators. We show that EBD injected into sick animals in the late stage of infection is rapidly sequestered in the viscera, while in healthy animals it remains within the plasma, causing the skin to turn a marked blue color. This test could be used in live animals to detect increased VP and to assess the ability of antiviral drugs and vasoactive compounds to prevent its onset. Finally, we describe a multiplexed assay to measure levels of serum factors during the course of Pichinde arenavirus infection and demonstrate that viremia and subsequent increase in white blood cell counts precede the elaboration of inflammatory mediators, which is followed by increased VP and death. CONCLUSIONS: This level of model characterization is essential to the evaluation of novel interventions designed to control the effects of virus-induced hypercytokinemia on host vascular function in VHF, which could lead to improved survival.


Assuntos
Permeabilidade Capilar/fisiologia , Azul Evans/farmacocinética , Febres Hemorrágicas Virais/patologia , Febres Hemorrágicas Virais/fisiopatologia , Vírus Pichinde/patogenicidade , Animais , Cricetinae , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Hematócrito , Mesocricetus , Albumina Sérica/análise
5.
Antimicrob Agents Chemother ; 53(5): 2120-8, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19273672

RESUMO

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.


Assuntos
Amantadina , Antivirais , Modelos Animais de Doenças , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Infecções por Orthomyxoviridae/tratamento farmacológico , Oseltamivir , Ribavirina , Amantadina/administração & dosagem , Amantadina/farmacologia , Amantadina/uso terapêutico , Animais , Antivirais/administração & dosagem , Antivirais/farmacologia , Antivirais/uso terapêutico , Linhagem Celular , Interações Medicamentosas , Farmacorresistência Viral/genética , Quimioterapia Combinada , Humanos , Virus da Influenza A Subtipo H5N1/genética , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana/métodos , Infecções por Orthomyxoviridae/virologia , Oseltamivir/administração & dosagem , Oseltamivir/farmacologia , Oseltamivir/uso terapêutico , Ribavirina/administração & dosagem , Ribavirina/farmacologia , Ribavirina/uso terapêutico , Organismos Livres de Patógenos Específicos , Resultado do Tratamento
6.
Antiviral Res ; 77(3): 215-24, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18222548

RESUMO

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.


Assuntos
Antivirais/uso terapêutico , Infecções por Bunyaviridae/tratamento farmacológico , Infecções por Bunyaviridae/prevenção & controle , Interferon Tipo I/uso terapêutico , Phlebovirus/efeitos dos fármacos , Alanina Transaminase/sangue , Animais , Antivirais/farmacologia , Infecções por Bunyaviridae/imunologia , Linhagem Celular , Sobrevivência Celular , Cricetinae , Feminino , Haplorrinos , Interferon Tipo I/farmacologia , Interferon-alfa , Icterícia/virologia , Fígado/virologia , Mesocricetus , Proteínas Recombinantes , Vírus da Febre do Vale do Rift/efeitos dos fármacos , Soro/virologia , Análise de Sobrevida , Resultado do Tratamento
7.
Int J Antimicrob Agents ; 31(4): 352-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18206353

RESUMO

The causes of death from intranasal cowpox virus infections in mice remain unclear. Hypotheses include severe pneumonitis, hepatitis and/or hyperproduction of cytokines and chemokines. This work explores these hypotheses by studying the influence of low- and high-volume virus inocula on viral pathogenesis. BALB/c mice were infected intranasally with a syncytium-forming variant of cowpox virus in 5 microL or 50 microL volumes containing the same infectious virus challenge dose. The 50 microL infection produced a more rapidly lethal disease associated with severe pneumonitis, high lung and nasal virus titres and increased cytokine and chemokine levels in the lungs and nasal tissue, whilst liver infection was minimal. The 5 microL inoculum infection was also lethal, but the infection was primarily confined to the upper respiratory tract and included elevated nasal cytokine and chemokine levels. Levels of the pro-inflammatory cytokine interleukin-6 were particularly high in both infections. Treatment of the infections with cidofovir (100mg/kg/day for 2 days starting 24h after virus exposure) led to survival and suppression of tissue virus titres. Treatment reduced pneumonitis in the 50 microL infection and lessened cytokine hyperproduction in both infections. We conclude that a 5 microL volume inoculum of cowpox virus causes a lethal upper respiratory tract infection, whilst the 50 microL inoculum targets both upper and lower respiratory tracts, with excessive release of systemic pro-inflammatory factors. Cidofovir effectively treated both infections and slowed viral replication sufficiently to subdue the exaggerated release of pro-inflammatory mediators.


Assuntos
Antivirais/uso terapêutico , Vírus da Varíola Bovina , Varíola Bovina/tratamento farmacológico , Varíola Bovina/patologia , Citosina/análogos & derivados , Organofosfonatos/uso terapêutico , Administração Intranasal , Animais , Peso Corporal/fisiologia , Quimiocinas/metabolismo , Cidofovir , Varíola Bovina/virologia , Citocinas/metabolismo , Citosina/uso terapêutico , Feminino , Células Gigantes/virologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Tamanho do Órgão
8.
Int Immunopharmacol ; 8(8): 1089-94, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18550012

RESUMO

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.


Assuntos
Antígenos de Protozoários/imunologia , Infecções por Bunyaviridae/imunologia , Infecções por Bunyaviridae/prevenção & controle , Eimeria/imunologia , Phlebovirus/imunologia , Animais , Antígenos de Protozoários/administração & dosagem , Infecções por Bunyaviridae/mortalidade , Cricetinae , Feminino , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Subunidade p40 da Interleucina-12/genética , Interleucina-18/administração & dosagem , Interleucina-18/imunologia , Interleucina-2/administração & dosagem , Interleucina-2/imunologia , Mesocricetus , Proibitinas , Proteínas Recombinantes/imunologia , Transcrição Gênica
9.
Antiviral Res ; 76(2): 124-9, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17658623

RESUMO

N-Methanocarbathymidine [(N)-MCT] is a newly identified inhibitor of orthopoxvirus replication in cell culture and in mice. Limited published animal studies indicated the compound is effective by intraperitoneal (i.p.) route at 10-100 mg/(kg day). More extensive studies using different treatment regimens in intranasally infected mice were conducted in order to further explore the potential of this compound compared to cidofovir in treating vaccinia virus infections. (N)-MCT was given twice a day for 7 days, whereas cidofovir was administered once a day for 2 days, each starting 24h after virus exposure for most experiments. (N)-MCT was not toxic up to 1000 mg/(kg day) by the i.p. treatment route. Oral and i.p. treatment regimens with (N)-MCT were directly compared during a vaccinia virus (IHD strain) infection, indicating that the nucleoside has good oral bioavailability in mice. Treatments by i.p. route with (N)-MCT (100 mg/(kg day)) reduced lung, nasal, and brain virus titers during an IHD virus infection, but not nearly to the same extent as i.p. cidofovir (100 mg/(kg day)). Treatment with both compounds decreased liver, spleen, and kidney virus titers, as well as reduced lung consolidation scores and lung weights. Onset of treatment could be delayed by 2 days with (N)-MCT and by 3 days with cidofovir, providing significant survival benefit during the IHD virus infection. Against a vaccinia virus (WR strain) infection in mice, i.p. (N)-MCT treatment prevented death at 500 mg/(kg day), which was comparable in activity to i.p. cidofovir (100 mg/(kg day)). Significant reductions in tissue virus titers occurred with both treatment regimens. (N)-MCT could be further pursued for its potential to treat orthopoxvirus infections in humans.


Assuntos
Antivirais/uso terapêutico , Timidina/análogos & derivados , Vaccinia virus/efeitos dos fármacos , Vacínia/tratamento farmacológico , Administração Oral , Animais , Cidofovir , Citosina/administração & dosagem , Citosina/análogos & derivados , Citosina/uso terapêutico , Injeções Intraperitoneais , Camundongos , Organofosfonatos/administração & dosagem , Organofosfonatos/uso terapêutico , Análise de Sobrevida , Timidina/administração & dosagem , Timidina/uso terapêutico , Timidina/toxicidade , Ensaio de Placa Viral
10.
Antiviral Res ; 73(1): 69-77, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16712967

RESUMO

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.


Assuntos
Antivirais/uso terapêutico , Varíola Bovina/tratamento farmacológico , Infecções Respiratórias/tratamento farmacológico , Timidina/análogos & derivados , Vacínia/tratamento farmacológico , Animais , Antivirais/farmacologia , Linhagem Celular , Varíola Bovina/virologia , Vírus da Varíola Bovina/efeitos dos fármacos , Vírus da Varíola Bovina/crescimento & desenvolvimento , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Coelhos , Infecções Respiratórias/virologia , Timidina/farmacologia , Timidina/uso terapêutico , Vacínia/virologia , Vaccinia virus/efeitos dos fármacos , Vaccinia virus/crescimento & desenvolvimento
11.
Antivir Chem Chemother ; 18(3): 125-32, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17626596

RESUMO

Avian influenza H5N1 infections can cause severe, lethal human infections. Whether influenza A virus treatments effectively ameliorate avian influenza H5N1 human infections is uncertain. The research objective was to evaluate the efficacy of novel zinc and other metallo-ion formulations in two influenza A mouse models. Mice infected with influenza A/Duck/MN/1525/81 (H5N1) virus were treated orally 48 h before virus exposure and then twice daily for 13 days with ZnAL42. The optimal dosing regimen for ZnAL42 was achieved at 17.28 mg/kg 48 h prior to virus exposure, twice daily for 7 days. The survival rate was 80% compared with 10% in the untreated control group and a 100% survival rate with ribavirin (75 mg/kg/day, twice a day for 5 days, beginning 4 h before virus exposure). ZnAL42 treatment significantly lessened the decline in arterial oxygen saturation (SaO2; P < 0.001). This regimen was also well tolerated by the mice. Manganese and selenium formulations were not inhibitory to virus replication when given therapeutically. Mice were also infected with influenza A/NWS/33 (H1N1) virus and were treated 48 h before virus exposure with three dosages of ZnAL42 (8.64, 1.46 or 0.24 mg/kg/day). Treatment was by oral gavage twice daily for 13 days. The highest dose of ZnAL42 was significantly inhibitory to the virus infection as seen by prevention of deaths and lessening of decline in SaO2. The data suggest that the prophylactic use of ZnAL42 is effective against avian influenza H5N1 or H1N1 virus infection in mice and should be further explored as an option for treating human influenza virus infections.


Assuntos
Compostos de Alumínio/farmacologia , Antivirais/farmacologia , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/prevenção & controle , Compostos de Zinco/farmacologia , Administração Oral , Animais , Antivirais/administração & dosagem , Modelos Animais de Doenças , Feminino , Humanos , Manganês/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Oxigênio/sangue , Ribavirina/administração & dosagem , Ribavirina/farmacologia , Selênio/farmacologia , Organismos Livres de Patógenos Específicos , Análise de Sobrevida
12.
Antiviral Res ; 69(3): 165-72, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16430975

RESUMO

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.


Assuntos
Adjuvantes Imunológicos , Infecções por Bunyaviridae/prevenção & controle , Infecções por Bunyaviridae/terapia , DNA Viral/imunologia , Lipossomos/imunologia , Phlebovirus/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Infecções por Bunyaviridae/imunologia , Infecções por Bunyaviridae/virologia , Citocinas/sangue , DNA Viral/administração & dosagem , Feminino , Injeções Intraperitoneais , Injeções Intravenosas , Lipossomos/administração & dosagem , Fígado/virologia , Camundongos , Análise de Sobrevida , Carga Viral
13.
Antivir Chem Chemother ; 17(4): 185-92, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17066897

RESUMO

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.


Assuntos
Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Infecções por Orthomyxoviridae/tratamento farmacológico , Oseltamivir/uso terapêutico , Ribavirina/uso terapêutico , Administração Oral , Animais , Antivirais/administração & dosagem , Antivirais/uso terapêutico , Células Cultivadas , Relação Dose-Resposta a Droga , Esquema de Medicação , Sinergismo Farmacológico , Quimioterapia Combinada , Camundongos , Oseltamivir/administração & dosagem , Placebos , Ribavirina/administração & dosagem , Fatores de Tempo
14.
Antivir Chem Chemother ; 17(4): 175-83, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17066896

RESUMO

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.


Assuntos
Infecções por Arenaviridae/tratamento farmacológico , Interferon Tipo I/uso terapêutico , Ribavirina/uso terapêutico , Doença Aguda/mortalidade , Administração Oral , Animais , Antivirais/uso terapêutico , Infecções por Arenaviridae/mortalidade , Cricetinae , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Feminino , Interferon Tipo I/administração & dosagem , Interferon-alfa , Placebos , Proteínas Recombinantes , Ribavirina/administração & dosagem , Análise de Sobrevida , Fatores de Tempo
15.
Int J Antimicrob Agents ; 23(5): 430-7, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15120719

RESUMO

Intranasal infection of BALB/c mice with the IHD strain of vaccinia virus was found to cause pneumonia, profound weight loss and death. Cidofovir, hexadecyloxypropyl-cidofovir (HDP-CDV), the diacetate ester prodrug of 2-amino-7-[(1,3-dihydroxy-2-propoxy)methyl]purine (HOE961), and ribavirin were used to treat the infections starting 24h after virus exposure. Single intraperitoneal (i.p.) cidofovir treatments of 100 and 30 mg/kg led to 90-100% survival compared with no survivors in the placebo group, whereas a 10 mg/kg dose was ineffective. The 100 mg/kg treatment reduced lung and snout virus titres on day 3 of the infection by 20- and 8-fold, respectively. Mean arterial oxygen saturation levels in these two cidofovir treatment groups were significantly higher than placebo on days 4 through 6 of the infection, indicating an improvement in lung function. Effects of cidofovir on viral pathogenesis were studied on days 1, 3 and 5 of the infection, and demonstrated statistically significant reductions in lung consolidation scores, lung weights, lung virus titre and snout virus titres on days 3 and 5. Cidofovir treatment also reduced virus titres in other tissues and body fluid, including blood, brain, heart, liver, salivary gland and spleen. HDP-CDV was given by oral gavage at 100, 50 and 25mg/kg doses one time only, resulting in 80-100% survival. Lower daily oral doses of 10 and 5mg/kg per day given for 5 days protected only 30% of animals from death. Oral doses (100, 50 and 25 mg/kg per day) of HOE961 for 5 days protected all animals, whereas equivalent oral doses of ribavirin were completely ineffective. The rapidity of recovery from weight loss during the infection was a function of dose of compound administered. These data indicate the utility of parenteral cidofovir, oral HDP-CDV and oral HOE961 in treating severe respiratory infections caused by this virus.


Assuntos
Antivirais/administração & dosagem , Antivirais/uso terapêutico , Citosina/análogos & derivados , Infecções Respiratórias/tratamento farmacológico , Infecções Respiratórias/virologia , Vacínia/tratamento farmacológico , Animais , Peso Corporal , Encéfalo/virologia , Cidofovir , Citosina/administração & dosagem , Citosina/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Coração/virologia , Fígado/virologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Organofosfonatos/administração & dosagem , Organofosfonatos/uso terapêutico , Oxigênio/sangue , Purinas/administração & dosagem , Purinas/uso terapêutico , Infecções Respiratórias/mortalidade , Infecções Respiratórias/patologia , Ribavirina/administração & dosagem , Ribavirina/uso terapêutico , Glândulas Salivares/virologia , Baço/virologia , Resultado do Tratamento , Vacínia/mortalidade , Vacínia/patologia , Vaccinia virus/isolamento & purificação , Ensaio de Placa Viral , Viremia
16.
Antivir Chem Chemother ; 15(5): 261-8, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15535048

RESUMO

Mannan, a polysaccharide preparation from Saccharomyces cerevisiae, has previously been shown to enhance influenza virus replication in mice by inhibiting host defense collectins. The use of mannan in infections may serve to broaden the types of influenza viruses that can be studied in rodent infection models. When mannan was co-administered with influenza B/Sichuan/379/99 virus to mice, the animals died from the infection, whereas mice infected with only virus survived. Three types of influenza A (H1N1) and another influenza B (Hong Kong/330/01) virus infection were also enhanced by mannan, but not four types of influenza A (H3N2) viruses. Mannan was used at 0.16 or 0.5 mg/mouse for optimal disease-enhancing activity using influenza B/Sichuan/379/99 virus. Using this model, influenza B/Sichuan/379/99 infections were treated with oseltamivir, ribavirin or viramidine (the carboxamidine derivative of ribavirin). When oral gavage treatments started 4 h before virus and mannan challenge, oseltamivir was effective at 2.5, 5 and 10 mg/kg/day. Ribavirin was active at 20, 40 and 80 mg/kg/day. Viramidine was effective at 80 and 160 mg/kg/day but not at 40 mg/kg/day. Active drug doses improved lung consolidation scores and lung weights, with decreases in lung virus titres also noted. Arterial oxygen saturation values in treated groups were significantly better than those of the placebo group on days 7-11 of the infection. Oseltamivir (5 mg/kg/day) and ribavirin (40 mg/kg/day) were used alone and in combination to determine how late after infection they could be beneficially administered. Ribavirin alone was very effective (90-100% survival of mice) when treatments started as late as 3 days after infection. Forty percent survival was evident even when treatments started 4 days post-infection. Oseltamivir was active starting treatments 1 day after virus exposure, but lost considerable efficacy when treatments began after that time. The combination of ribavirin and oseltamivir appeared to be no better than ribavirin alone, due to the stronger beneficial effect of ribavirin in this model. The overall results demonstrate that mannan can be used to enhance certain non-lethal influenza virus infections sufficiently to allow antiviral studies.


Assuntos
Acetamidas/uso terapêutico , Vírus da Influenza B/efeitos dos fármacos , Infecções por Orthomyxoviridae/tratamento farmacológico , Ribavirina/análogos & derivados , Ribavirina/uso terapêutico , Administração Oral , Animais , Antivirais/uso terapêutico , Relação Dose-Resposta a Droga , Hong Kong , Pulmão/efeitos dos fármacos , Pulmão/virologia , Mananas/farmacologia , Camundongos , Tamanho do Órgão/efeitos dos fármacos , Oseltamivir , Taxa de Sobrevida
17.
Antivir Chem Chemother ; 23(4): 151-9, 2014 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-23337126

RESUMO

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.


Assuntos
Antivirais/química , Antivirais/uso terapêutico , Arenavirus do Novo Mundo/efeitos dos fármacos , Febre Hemorrágica Americana/tratamento farmacológico , Animais , Chlorocebus aethiops , Humanos , Vírus Junin/efeitos dos fármacos , Camundongos , Fenóis/química , Fenóis/uso terapêutico , Sulfetos/química , Sulfetos/uso terapêutico , Células Vero
18.
PLoS One ; 8(7): e68685, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874722

RESUMO

An adenovirus 5 vector encoding for mouse interferon alpha, subtype 5 (mDEF201) was evaluated for efficacy against lethal cowpox (Brighton strain) and vaccinia (WR strain) virus respiratory and systemic infections in mice. Two routes of mDEF201 administration were used, nasal sinus (5-µl) and pulmonary (50-µl), to compare differences in efficacy, since the preferred treatment of humans would be in a relatively small volume delivered intranasally. Lower respiratory infections (LRI), upper respiratory infections (URI), and systemic infections were induced by 50-µl intranasal, 10-µl intranasal, and 100-µl intraperitoneal virus challenges, respectively. mDEF201 treatments were given prophylactically either 24 h (short term) or 56d (long-term) prior to virus challenge. Single nasal sinus treatments of 10(6) and 10(7) PFU/mouse of mDEF201 protected all mice from vaccinia-induced LRI mortality (comparable to published studies with pulmonary delivered mDEF201). Systemic vaccinia infections responded significantly better to nasal sinus delivered mDEF201 than to pulmonary treatments. Cowpox LRI infections responded to 10(7) mDEF201 treatments, but a 10(6) dose was only weakly protective. Cowpox URI infections were equally treatable by nasal sinus and pulmonary delivered mDEF201 at 10(7) PFU/mouse. Dose-responsive prophylaxis with mDEF201, given one time only 56 d prior to initiating a vaccinia virus LRI infection, was 100% protective from 10(5) to 10(7) PFU/mouse. Improvements in lung hemorrhage score and lung weight were evident, as were decreases in liver, lung, and spleen virus titers. Thus, mDEF201 was able to treat different vaccinia and cowpox virus infections using both nasal sinus and pulmonary treatment regimens, supporting its development for humans.


Assuntos
Vírus da Varíola Bovina/imunologia , Varíola Bovina/prevenção & controle , Vetores Genéticos/genética , Interferons/genética , Vaccinia virus/imunologia , Vacínia/prevenção & controle , Adenoviridae/genética , Administração Intranasal , Animais , Varíola Bovina/mortalidade , Modelos Animais de Doenças , Feminino , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intraperitoneais , Camundongos , Vacínia/mortalidade
19.
Antivir Chem Chemother ; 21(6): 231-7, 2011 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-21730370

RESUMO

BACKGROUND: Limited in vivo studies in the scientific literature suggest that components of green tea and elderberry may be beneficial in treating influenza virus infections. They are thought to act by blocking virus adsorption to cells. TheraMax(®) is a proprietary medication administered by nasal spray that contains both green tea and elderberry extracts that was evaluated for antiviral activity. METHODS: TheraMax was tested by dilution in Madin-Darby canine kidney cell cultures in standard viral cytopathic effect (CPE) inhibition and virucidal assays against eight influenza A and B strains. It was also administered intranasally to mice to determine protective activity compared to oral oseltamivir against an influenza A/NWS/33 (H1N1) infection. RESULTS: In cell culture assays, TheraMax was found to inhibit viral CPE by 50% at a 1:20 dilution for seven of the eight virus strains, with no virucidal activity at 1:2 dilution. The undiluted product was administered to anaesthetized mice twice daily for 4 days starting 2 h before or 12 h after infection. Alternatively, TheraMax and virus were combined for treatment and infection. Oseltamivir was given orally twice daily for 5 days at 10 mg/kg/day starting at -2 h. TheraMax (combined directly with virus) and oseltamivir each prevented death and curtailed weight loss during the infection, and improved lung haemorrhage scores on day 6. TheraMax starting at -2 h or 12 h significantly delayed death by >2 days and reduced lung haemorrhage scores, but most animals died. CONCLUSIONS: These combined results indicate that TheraMax delayed symptoms during animal infections, likely through blocking of virus adsorption.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Infecções por Orthomyxoviridae/tratamento farmacológico , Administração Intranasal , Animais , Antivirais/administração & dosagem , Camellia sinensis/química , Linhagem Celular , Cães , Avaliação Pré-Clínica de Medicamentos , Hemorragia/patologia , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pneumopatias/patologia , Camundongos , Camundongos Endogâmicos BALB C , Cavidade Nasal/efeitos dos fármacos , Cavidade Nasal/patologia , Oseltamivir/administração & dosagem , Oseltamivir/farmacologia , Extratos Vegetais/farmacologia , Sambucus/química , Testes de Toxicidade , Redução de Peso/efeitos dos fármacos
20.
PLoS One ; 6(10): e26330, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22022603

RESUMO

An adenovirus 5 vector encoding for mouse interferon alpha, subtype 5 (mDEF201) was evaluated for efficacy against lethal vaccinia virus (WR strain) respiratory infections in mice. mDEF201 was administered as a single intranasal treatment either prophylactically or therapeutically at doses of 10(6) to 10(8) plaque forming units/mouse. When the prophylactic treatment was given at 56 days prior to infection, it protected 90% of animals from death (100% protection for treatments given between 1-49 days pre-infection), with minimal weight loss occurring during infection. Surviving animals re-challenged with virus 22 days after the primary infection were protected from death, indicating that mDEF201 did not compromise the immune response against the initial infection. Post-exposure therapy was given between 6-24 h after vaccinia virus exposure and protection was afforded by a 10(8) dose of mDEF201 given at 24 h, whereas a 10(7) dose was effective up to 12 h. Comparisons were made of the ability of mDEF201, given either 28 or 1 day prior to infection, to inhibit tissue virus titers and lung infection parameters. Lung, liver, and spleen virus titers were inhibited to nearly the same extent by either treatment, as were lung weights and lung hemorrhage scores (indicators of pneumonitis). Lung virus titers were significantly (>100-fold) lower than in the placebo group, and the other infection parameters in mDEF201 treated mice were nearly at baseline. In contrast, viral titers and lung infection parameters were high in the placebo group on day 5 of the infection. These results demonstrate the long-acting prophylactic and treatment capacity of mDEF201 to combat vaccinia virus infections.


Assuntos
Adenoviridae/genética , Vetores Genéticos/genética , Interferon-alfa/uso terapêutico , Infecções Respiratórias/tratamento farmacológico , Vaccinia virus/fisiologia , Vacínia/tratamento farmacológico , Vacínia/prevenção & controle , Administração Intranasal , Animais , Peso Corporal/efeitos dos fármacos , Cidofovir , Citosina/análogos & derivados , Citosina/farmacologia , Citosina/uso terapêutico , Feminino , Interferon-alfa/administração & dosagem , Interferon-alfa/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Organofosfonatos/farmacologia , Organofosfonatos/uso terapêutico , Substâncias Protetoras/farmacologia , Substâncias Protetoras/uso terapêutico , Infecções Respiratórias/patologia , Infecções Respiratórias/prevenção & controle , Infecções Respiratórias/virologia , Análise de Sobrevida , Fatores de Tempo , Vacínia/patologia , Vacínia/virologia , Vaccinia virus/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA