Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Antiviral Res ; 149: 1-6, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29113740

RESUMO

Pritelivir, a helicase-primase inhibitor, has excellent in vitro and in vivo activity against human herpes simplex virus (HSV). Mice lethally infected with HSV type 1 or 2, including acyclovir-resistant strains, were treated 72 h after infection for 7 days with pritelivir or acyclovir. Both drugs were administered orally twice daily either alone or in combination. Dosages of pritelivir from 0.3 to 30 mg/kg reduced mortality (P < 0.001) against HSV-1, E-377. With an acyclovir resistant HSV-1, 11360, pritelivir at 1 and 3 mg/kg increased survival (P < 0.005). With HSV-2, MS infected mice, all dosages higher than the 0.3 mg/kg dose of pritelivir were effective (P < 0.005). For acyclovir resistant HSV-2, strain 12247, pritelivir dosages of 1-3 mg/kg significantly improved survival (P < 0.0001). Combination therapies of pritelivir at 0.1 or 0.3 mg/kg/dose with acyclovir (10 mg/kg/dose) were protective (P < 0.0001) when compared to the vehicle treated group against HSV-2, strain MS (in line with previous data using HSV-1). An increased mean days to death (P < 0.05) was also observed and was indicative of a potential synergy. Pharmacokinetic studies were performed to determine pritelivir concentrations and a dose dependent relationship was found in both plasma and brain samples regardless of infection status or time of initiation of dosing. In summary, pritelivir was shown to be active when treatment was delayed to 72 h post viral inoculation and appeared to synergistically inhibit mortality in this model in combination with acyclovir. We conclude pritelivir has potent and resistance-breaking antiviral efficacy with potential for the treatment of potentially life-threatening HSV type 1 and 2 infections, including herpes simplex encephalitis.


Assuntos
Aciclovir/farmacologia , Antivirais/farmacologia , Encefalite por Herpes Simples/virologia , Piridinas/farmacologia , Tiazóis/farmacologia , Aciclovir/administração & dosagem , Aciclovir/farmacocinética , Animais , Antivirais/administração & dosagem , Antivirais/farmacocinética , Modelos Animais de Doenças , Quimioterapia Combinada , Encefalite por Herpes Simples/tratamento farmacológico , Encefalite por Herpes Simples/mortalidade , Encefalite por Herpes Simples/patologia , Feminino , Humanos , Camundongos , Piridinas/administração & dosagem , Piridinas/farmacocinética , Sulfonamidas , Tiazóis/administração & dosagem , Tiazóis/farmacocinética , Distribuição Tecidual , Resultado do Tratamento
2.
Sci Transl Med ; 6(265): 265ra169, 2014 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-25473037

RESUMO

Herpesviruses are highly prevalent and maintain lifelong latent reservoirs, thus posing challenges to the control of herpetic disease despite the availability of antiviral pharmaceuticals that target viral DNA replication. The initiation of herpes simplex virus infection and reactivation from latency is dependent on a transcriptional coactivator complex that contains two required histone demethylases, LSD1 (lysine-specific demethylase 1) and a member of the JMJD2 family (Jumonji C domain-containing protein 2). Inhibition of either of these enzymes results in heterochromatic suppression of the viral genome and blocks infection and reactivation in vitro. We demonstrate that viral infection can be epigenetically suppressed in three animal models of herpes simplex virus infection and disease. Treating animals with the monoamine oxidase inhibitor tranylcypromine to inhibit LSD1 suppressed viral lytic infection, subclinical shedding, and reactivation from latency in vivo. This phenotypic suppression was correlated with enhanced epigenetic suppression of the viral genome and suggests that, even during latency, the chromatin state of the virus is dynamic. Therefore, epi-pharmaceuticals may represent a promising approach to treat herpetic diseases.


Assuntos
Epigênese Genética , Infecções por Herpesviridae/metabolismo , Oxirredutases N-Desmetilantes/antagonistas & inibidores , Oxirredutases N-Desmetilantes/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Genoma Viral , Cobaias , Histona Desmetilases , Camundongos , Camundongos Endogâmicos BALB C , Inibidores da Monoaminoxidase/química , Fenótipo , Estrutura Terciária de Proteína , Coelhos , Recidiva , Tranilcipromina/química , Vagina/virologia , Ativação Viral , Latência Viral , Replicação Viral/efeitos dos fármacos , Eliminação de Partículas Virais
3.
Int Trends Immun ; 2(2): 87-92, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25215309

RESUMO

Natural serine protease inhibitors (serpins) elicit sensing of a microbial cell intruder and activate an intrinsic cellular immune response in HIV and HCV infected cells. Here, we demonstrate in vitro inhibition of HSV with serpin antithrombin III (ATIII) early during infection pointing towards inhibition of an entry event. We also found reduction of mortality from 90% to 40% in an abrasion mice model demonstrating a strong reduction of infection in vivo. Our data also indicated that this treatment might be suitable for drug-resistant viruses since high inhibition of an acyclovir-resistant HSV-1 strain was found. Thus, an ATIII tropical treatment might be used for immunocompromised patients where prolonged treatment leads to drug resistant HSV-1 strains. Understanding how ATIII regulates HSV-1 infections may reveal new avenues for therapeutic interventions.

4.
J Virol ; 86(9): 5304-13, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22379082

RESUMO

Herpes simplex virus 1 (HSV-1) mutants that lack the γ(1)34.5 gene are unable to replicate in the central nervous system but maintain replication competence in dividing cell populations, such as those found in brain tumors. We have previously demonstrated that a γ(1)34.5-deleted HSV-1 expressing murine interleukin-12 (IL-12; M002) prolonged survival of immunocompetent mice in intracranial models of brain tumors. We hypothesized that M002 would be suitable for use in clinical trials for patients with malignant glioma. To test this hypothesis, we (i) compared the efficacy of M002 to three other HSV-1 mutants, R3659, R8306, and G207, in murine models of brain tumors, (ii) examined the safety and biodistribution of M002 in the HSV-1-sensitive primate Aotus nancymae following intracerebral inoculation, and (iii) determined whether murine IL-12 produced by M002 was capable of activating primate lymphocytes. Results are summarized as follows: (i) M002 demonstrated superior antitumor activity in two different murine brain tumor models compared to three other genetically engineered HSV-1 mutants; (ii) no significant clinical or magnetic resonance imaging evidence of toxicity was observed following direct inoculation of M002 into the right frontal lobes of A. nancymae; (iii) there was no histopathologic evidence of disease in A. nancymae 1 month or 5.5 years following direct inoculation; and (iv) murine IL-12 produced by M002 activates A. nancymae lymphocytes in vitro. We conclude that the safety and preclinical efficacy of M002 warrants the advancement of a Δγ(1)34.5 virus expressing IL-12 to phase I clinical trials for patients with recurrent malignant glioma.


Assuntos
Vetores Genéticos/genética , Interleucina-12/genética , Simplexvirus/genética , Aciclovir/farmacologia , Animais , Antivirais/farmacologia , Aotidae , Encéfalo/patologia , Linhagem Celular , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos , Feminino , Expressão Gênica , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/efeitos adversos , Glioma/genética , Glioma/mortalidade , Glioma/terapia , Humanos , Interleucina-12/metabolismo , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos SCID , Simplexvirus/efeitos dos fármacos , Análise de Sobrevida , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Bioorg Med Chem ; 20(8): 2669-74, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22417649

RESUMO

Synthesis of 6-deoxycyclopropavir (10), a prodrug of cyclopropavir (1) and its in vitro and in vivo antiviral activity is described. 2-Amino-6-chloropurine methylenecyclopropane 13 was transformed to its 6-iodo derivative 14 which was reduced to prodrug 10. It is converted to cyclopropavir (1) by the action of xanthine oxidase and this reaction can also occur in vivo. Compound 10 lacked significant in vitro activity against human cytomegalovirus (HCMV), human herpes virus 1 and 2 (HSV-1 and HSV-2), human immunodeficiency virus type 1 (HIV-1), human hepatitis B virus (HBV), Epstein-Barr virus (EBV), vaccinia virus and cowpox virus. In contrast, prodrug 10 given orally was as active as cyclopropavir (1) reported previously [Kern, E. R.; Bidanset, D. J.; Hartline, C. B.; Yan, Z.; Zemlicka, J.; Quenelle, D. C. et al. Antimicrob. Agents Chemother. 2004, 48, 4745] against murine cytomegalovirus (MCMV) infection in mice and against HCMV in severe combined immunodeficient (SCID) mice.


Assuntos
Antivirais/síntese química , Antivirais/farmacologia , Ciclopropanos/síntese química , Ciclopropanos/farmacologia , Guanina/análogos & derivados , Pró-Fármacos/síntese química , Pró-Fármacos/farmacologia , Administração Oral , Animais , Antivirais/administração & dosagem , Vírus da Varíola Bovina/efeitos dos fármacos , Ciclopropanos/química , Citomegalovirus/efeitos dos fármacos , Guanina/síntese química , Guanina/química , Guanina/farmacologia , HIV-1/efeitos dos fármacos , Vírus da Hepatite B/efeitos dos fármacos , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Herpesvirus Humano 4/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Testes de Sensibilidade Microbiana , Estrutura Molecular , Pró-Fármacos/administração & dosagem , Vaccinia virus/efeitos dos fármacos
6.
Antivir Chem Chemother ; 22(3): 131-7, 2011 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-22095521

RESUMO

BACKGROUND: N-methanocarbathymidine (N-MCT) has previously been shown to be effective against lethal orthopoxvirus and herpes simplex virus type-1 infections in mice. In this investigation, the antiviral activity of N-MCT was assessed against herpes simplex virus type-2 (HSV-2) in BALB/c mice. METHODS: BALB/c mice were infected intranasally with a lethal challenge dose of HSV-2. N-MCT was administered orally twice daily to mice using doses of 0.01 to 100 mg/kg to determine effects on survival and on viral replication in organ and central nervous system (CNS) samples. RESULTS: N-MCT provided significant protection from mortality even when treatments were delayed until 3 days after viral infection. Viral replication in organ and CNS samples from N-MCT-treated mice was reduced below the limit of detection after 4 days of treatment. CONCLUSIONS: These results indicated that low dose N-MCT treatment was more effective than acyclovir therapy. N-MCT may be effective against HSV disease in humans and is currently undergoing preclinical evaluation. In particular, its potential use as a combination therapy for HSV, with its differing metabolism from acyclovir, make it a promising compound to develop for human use.


Assuntos
Antivirais/administração & dosagem , Antivirais/farmacologia , Herpes Simples/tratamento farmacológico , Herpesvirus Humano 2/efeitos dos fármacos , Timidina/análogos & derivados , Administração Oral , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Herpes Simples/virologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Taxa de Sobrevida , Timidina/administração & dosagem , Timidina/farmacologia
7.
Antimicrob Agents Chemother ; 55(10): 4728-34, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21788472

RESUMO

Although acyclovir (ACV) has proven to be of value in the therapy of certain herpes simplex virus (HSV) infections, there is a need for more effective therapies, particularly for serious infections in neonates and immunocompromised individuals, where resistance to this drug can be problematic. CMX001 is an orally bioavailable lipid conjugate of cidofovir that is substantially less nephrotoxic than the parent drug and has excellent antiviral activity against all the human herpesviruses. This compound retains full antiviral activity against ACV-resistant laboratory and clinical isolates. The combined efficacy of CMX001 and ACV was evaluated in a new real-time PCR combination assay, which demonstrated that the combination synergistically inhibited the replication of HSV in cell culture. This was also confirmed in murine models of HSV infection, where the combined therapy with these two drugs synergistically reduced mortality. These results suggest that CMX001 may be effective in the treatment of ACV-resistant HSV infections and as an adjunct therapy in individuals with suboptimal responses to ACV.


Assuntos
Aciclovir/farmacologia , Antivirais/farmacologia , Citosina/análogos & derivados , Herpes Simples/tratamento farmacológico , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Organofosfonatos/farmacologia , Aciclovir/uso terapêutico , Aciclovir/toxicidade , Animais , Antivirais/uso terapêutico , Antivirais/toxicidade , Células Cultivadas , Citosina/farmacologia , Citosina/uso terapêutico , Citosina/toxicidade , Farmacorresistência Viral , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Herpes Simples/virologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Organofosfonatos/uso terapêutico , Organofosfonatos/toxicidade
8.
J Infect Dis ; 202(10): 1492-9, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20923374

RESUMO

CMX001, an orally active lipid conjugate of cidofovir, is 50 times more active in vitro against herpes simplex virus (HSV) replication than acyclovir or cidofovir. These studies compared the efficacy of CMX001 to acyclovir in BALB/c mice inoculated intranasally with HSV types 1 or 2. CMX001 was effective in reducing mortality using doses of 5 to 1.25 mg/kg administered orally once daily, even when treatments were delayed 48-72 h post viral inoculation. Organ samples obtained from mice treated with CMX001 had titers 3-5 log(10) plaque-forming units per gram of tissue lower than samples obtained from mice treated with acyclovir, including 5 different regions of the brain. Detectable concentrations of drug-related radioactivity were documented in the central nervous system of mice after oral administration of (14)C-CMX001. These studies indicate that CMX001 penetrates the blood-brain barrier, is a potent inhibitor of HSV replication in disseminated infections and central nervous system infections, and is superior to acyclovir.


Assuntos
Antivirais/administração & dosagem , Antivirais/farmacocinética , Citosina/análogos & derivados , Herpes Simples/tratamento farmacológico , Herpesvirus Humano 1 , Herpesvirus Humano 2 , Organofosfonatos/administração & dosagem , Organofosfonatos/farmacocinética , Administração Oral , Animais , Citosina/administração & dosagem , Citosina/farmacocinética , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Distribuição Tecidual
9.
Viruses ; 2(12): 2681-95, 2010 12.
Artigo em Inglês | MEDLINE | ID: mdl-21994637

RESUMO

Although a large number of compounds have been identified with antiviral activity against orthopoxviruses in tissue culture systems, it is highly preferred that these compounds have activity in vivo before they can be seriously considered for further development. One of the most commonly used animal models for the confirmation of this activity has been the use of mice infected with either vaccinia or cowpox viruses. These model systems have the advantage that they are relatively inexpensive, readily available and do not require any special containment facilities; therefore, relatively large numbers of compounds can be evaluated in vivo for their activity. The two antiviral agents that have progressed from preclinical studies to human safety trials for the treatment of orthopoxvirus infections are the cidofovir analog, CMX001, and an inhibitor of extracellular virus formation, ST-246. These compounds are the ones most likely to be used in the event of a bioterror attack. The purpose of this communication is to review the advantages and disadvantages of using mice infected with vaccinia and cowpox virus as surrogate models for human orthopoxvirus infections and to summarize the activity of CMX001 and ST-246 in these model infections.

10.
Antimicrob Agents Chemother ; 53(12): 5251-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19770274

RESUMO

A series of 4'-thionucleosides were synthesized and evaluated for activities against orthopoxviruses and herpesviruses. We reported previously that one analog, 5-iodo-4'-thio-2'-deoxyuridine (4'-thioIDU), exhibits good activity both in vitro and in vivo against two orthopoxviruses. This compound also has good activity in cell culture against many of the herpesviruses. It inhibited the replication of herpes simplex virus type 1 (HSV-1), HSV-2, and varicella-zoster virus with 50% effective concentrations (EC(50)s) of 0.1, 0.5, and 2 microM, respectively. It also inhibited the replication of human cytomegalovirus (HCMV) with an EC(50) of 5.9 microM but did not selectively inhibit Epstein-Barr virus, human herpesvirus 6, or human herpesvirus 8. While acyclovir-resistant strains of HSV-1 and HSV-2 were comparatively resistant to 4'-thioIDU, it retained modest activity (EC(50)s of 4 to 12 microM) against these strains. Some ganciclovir-resistant strains of HCMV also exhibited reduced susceptibilities to the compound, which appeared to be related to the specific mutations in the DNA polymerase, consistent with the observed incorporation of the compound into viral DNA. The activity of 4'-thioIDU was also evaluated using mice infected intranasally with the MS strain of HSV-2. Although there was no decrease in final mortality rates, the mean length of survival after inoculation increased significantly (P < 0.05) for all animals receiving 4'-thioIDU. The findings from the studies presented here suggest that 4'-thioIDU is a good inhibitor of some herpesviruses, as well as orthopoxviruses, and this class of compounds warrants further study as a therapy for infections with these viruses.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , Infecções por Herpesviridae/tratamento farmacológico , Herpesviridae/efeitos dos fármacos , Nucleosídeos de Pirimidina/farmacologia , Nucleosídeos de Pirimidina/uso terapêutico , Replicação Viral/efeitos dos fármacos , Animais , Antivirais/efeitos adversos , Antivirais/química , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/fisiologia , Farmacorresistência Viral/efeitos dos fármacos , Farmacorresistência Viral/genética , Técnica Indireta de Fluorescência para Anticorpo , Herpesviridae/genética , Infecções por Herpesviridae/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/genética , Herpesvirus Humano 2/efeitos dos fármacos , Herpesvirus Humano 2/genética , Herpesvirus Humano 6/efeitos dos fármacos , Herpesvirus Humano 6/genética , Herpesvirus Humano 8/efeitos dos fármacos , Herpesvirus Humano 8/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Estrutura Molecular , Nucleosídeos de Pirimidina/síntese química , Nucleosídeos de Pirimidina/química , Proteínas Virais/genética , Proteínas Virais/fisiologia
11.
Antimicrob Agents Chemother ; 53(2): 572-9, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19029322

RESUMO

As part of a program to identify new compounds that have activity against orthopoxviruses, a number of 4'-thionucleosides were synthesized and evaluated for their efficacies against vaccinia and cowpox viruses. Seven compounds that were active at about 1 microM against both viruses in human cells but that did not have significant toxicity were identified. The 5-iodo analog, 1-(2-deoxy-4-thio-beta-d-ribofuranosyl)-5-iodouracil (4'-thioIDU), was selected as a representative molecule; and this compound also inhibited viral DNA synthesis at less than 1 microM but only partially inhibited the replication of a recombinant vaccinia virus that lacked a thymidine kinase. This compound retained complete activity against cidofovir- and ST-246-resistant mutants. To determine if this analog had activity in an animal model, mice were infected intranasally with vaccinia or cowpox virus and treatment with 4'-thioIDU was given intraperitoneally or orally twice daily at 50, 15, 5, or 1.5 mg/kg of body weight beginning at 24 to 120 h postinfection and was continued for 5 days. Almost complete protection (87%) was observed when treatment with 1.5 mg/kg was begun at 72 h postinfection, and significant protection (73%) was still obtained when treatment with 5 mg/kg was initiated at 96 h. Virus titers in the liver, spleen, and kidney were reduced by about 4 log(10) units and about 2 log(10) units in mice infected with vaccinia virus and cowpox virus, respectively. These results indicate that 4'-thioIDU is a potent, nontoxic inhibitor of orthopoxvirus replication in cell culture and experimental animal infections and suggest that it may have potential for use in the treatment of orthopoxvirus infections in animals and humans.


Assuntos
Antivirais/uso terapêutico , Nucleosídeos/uso terapêutico , Orthopoxvirus , Infecções por Poxviridae/tratamento farmacológico , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Varíola Bovina/tratamento farmacológico , Efeito Citopatogênico Viral/efeitos dos fármacos , DNA Viral/biossíntese , DNA Viral/genética , Farmacorresistência Viral , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Poxviridae/virologia , Relação Estrutura-Atividade , Vacínia/tratamento farmacológico , beta-Galactosidase/metabolismo
12.
Antiviral Res ; 80(2): 223-4, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18573279

RESUMO

These studies were performed to determine the effect of AD-472, an attenuated human herpes simplex virus (HSV) type 2 or HSV-2 glycoprotein D (gD) when combined with an adjuvant, GPI-0100, a semi-synthetic Quillaja Saponin analog in a genital HSV-2 infection in guinea pigs. While animals immunized with either vaccine had reduced clinical disease, GPI-0100 only improved the efficacy of gD and did not affect the efficacy of the live vaccine. Neither vaccine had any therapeutic effect if administered 24 h after viral infection.


Assuntos
Adjuvantes Imunológicos , Herpes Genital/prevenção & controle , Vacinas contra o Vírus do Herpes Simples/imunologia , Herpesvirus Humano 2/imunologia , Saponinas/imunologia , Vacinação , Vacinas Atenuadas/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Feminino , Cobaias , Herpes Genital/imunologia , Humanos
13.
Virol J ; 5: 58, 2008 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-18479513

RESUMO

BACKGROUND: The emergence of drug resistant viruses, together with the possibility of increased virulence, is an important concern in the development of new antiviral compounds. Cidofovir (CDV) is a phosphonate nucleotide that is approved for use against cytomegalovirus retinitis and for the emergency treatment of smallpox or complications following vaccination. One mode of action for CDV has been demonstrated to be the inhibition of the viral DNA polymerase. RESULTS: We have isolated several CDV resistant (CDVR) vaccinia viruses through a one step process, two of which have unique single mutations within the DNA polymerase. An additional resistant virus isolate provides evidence of a second site mutation within the genome involved in CDV resistance. The CDVR viruses were 3-7 fold more resistant to the drug than the parental viruses. The virulence of the CDVR viruses was tested in mice inoculated intranasally and all were found to be attenuated. CONCLUSION: Resistance to CDV in vaccinia virus can be conferred individually by at least two different mutations within the DNA polymerase gene. Additional genes may be involved. This one step approach for isolating resistant viruses without serial passage and in the presence of low doses of drug minimizes unintended secondary mutations and is applicable to other potential antiviral agents.


Assuntos
Citosina/análogos & derivados , Farmacorresistência Viral , Organofosfonatos/farmacologia , Vaccinia virus/efeitos dos fármacos , Vaccinia virus/isolamento & purificação , Animais , Antivirais/farmacologia , Linhagem Celular , Chlorocebus aethiops , Cidofovir , Citosina/farmacologia , DNA Polimerase Dirigida por DNA/química , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Feminino , Humanos , Camundongos , Modelos Moleculares , Mutação , Vacínia/mortalidade , Vacínia/virologia , Vaccinia virus/patogenicidade , Vaccinia virus/fisiologia , Células Vero , Ensaio de Placa Viral , Virulência
14.
Virol J ; 5: 39, 2008 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-18321387

RESUMO

BACKGROUND: The vaccinia virus (VV) F2L gene encodes a functional deoxyuridine triphosphatase (dUTPase) that catalyzes the conversion of dUTP to dUMP and is thought to minimize the incorporation of deoxyuridine residues into the viral genome. Previous studies with with a complex, multigene deletion in this virus suggested that the gene was not required for viral replication, but the impact of deleting this gene alone has not been determined in vitro or in vivo. Although the crystal structure for this enzyme has been determined, its potential as a target for antiviral therapy is unclear. RESULTS: The F2L gene was replaced with GFP in the WR strain of VV to assess its effect on viral replication. The resulting virus replicated well in cell culture and its replication kinetics were almost indistinguishable from those of the wt virus and attained similar titers. The virus also appeared to be as pathogenic as the WR strain suggesting that it also replicated well in mice. Cells infected with the dUTPase mutant would be predicted to affect pyrimidine deoxynucleotide pools and might be expected to exhibit altered susceptibility to pyrimidine analogs. The antiviral activity of cidofovir and four thymidine analogs were evaluated both in the mutant and the parent strain of this virus. The dUTPase knockout remained fully susceptible to cidofovir and idoxuridine, but was hypersensitive to the drug (N)-methanocarbathymidine, suggesting that pyrimidine metabolism was altered in cells infected with the mutant virus. The absence of dUTPase should reduce cellular dUMP pools and may result in a reduced conversion to dTMP by thymidylate synthetase or an increased reliance on the salvage of thymidine by the viral thymidine kinase. CONCLUSION: We confirmed that F2L was not required for replication in cell culture and determined that it does not play a significant role on virulence of the virus in intranasally infected mice. The recombinant virus is hypersensitive to (N)-methanocarbathymidine and may reflect metabolic differences in the mutant virus.


Assuntos
Antivirais/farmacologia , Pirofosfatases/metabolismo , Timidina/análogos & derivados , Vaccinia virus/fisiologia , Vacínia/virologia , Replicação Viral/efeitos dos fármacos , Animais , Feminino , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Pirimidinas/metabolismo , Pirofosfatases/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Timidina/farmacologia , Vaccinia virus/efeitos dos fármacos , Vaccinia virus/enzimologia , Vaccinia virus/patogenicidade , Ensaio de Placa Viral , Virulência
15.
Antiviral Res ; 79(2): 133-5, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18336926

RESUMO

We utilized BALB/c mice infected with murine CMV (MCMV) or severe combined immunodeficient (SCID) mice implanted with human fetal tissue and infected with HCMV to determine the efficacy of (S)-9-[3-hydroxy-2-(phophonomethoxy)propyl]adenine ((S)-HPMPA), hexadecyloxypropyl-(S)-HPMPA (HDP-(S)-HPMPA) or octadecyloxyethyl-(S)-HPMPA (ODE-(S)-HPMPA). In MCMV-infected BALB/c mice, oral HDP-(S)-HPMPA at 30 mg/kg significantly reduced mortality when started 24-48 h post inoculation. In the experimental HCMV infection, oral administration of vehicle or 10mg/kg of (S)-HPMPA, HDP-(S)-HPMPA or ODE-(S)-HPMPA was initiated 24h after infection and continued for 28 consecutive days. Cidofovir (CDV), at 20mg/kg given i.p., was used as a positive control. HDP-(S)-HPMPA or ODE-(S)-HPMPA significantly reduced viral replication compared to vehicle-treated mice, while oral (S)-HPMPA was ineffective.


Assuntos
Adenina/análogos & derivados , Antivirais/uso terapêutico , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Herpesviridae/tratamento farmacológico , Organofosfonatos/uso terapêutico , Adenina/administração & dosagem , Adenina/farmacologia , Adenina/uso terapêutico , Administração Oral , Animais , Antivirais/administração & dosagem , Antivirais/farmacologia , Cidofovir , Citomegalovirus/efeitos dos fármacos , Citosina/administração & dosagem , Citosina/análogos & derivados , Citosina/uso terapêutico , Fígado/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Muromegalovirus/efeitos dos fármacos , Organofosfonatos/administração & dosagem , Organofosfonatos/farmacologia , Análise de Sobrevida
16.
Antimicrob Agents Chemother ; 51(11): 3940-7, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17846137

RESUMO

We have previously reported that (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine, or (S)-HPMPA, is active in vitro against cowpox virus (CV) and vaccinia virus (VV) but is not active orally in animals. However, the ether lipid esters of (S)-HPMPA, hexadecyloxypropyl-[(S)-HPMPA] [HDP-(S)-HPMPA] and octadecyloxyethyl-[(S)-HPMPA] [ODE-(S)-HPMPA], had significantly enhanced activity in vitro and are orally bioavailable in mice. In the current study, HDP-(S)-HPMPA and ODE-(S)-HPMPA were prepared in water and administered once daily by oral gavage to mice at doses of 30, 10, and 3 mg/kg of body weight for 5 days beginning 24, 48, or 72 h after inoculation with CV or VV. Oral HDP-(S)-HPMPA and ODE-(S)-HPMPA were both highly effective (P < 0.001) at preventing mortality due to CV at 30 mg/kg, even when treatments were delayed until up to 72 h postinfection. ODE-(S)-HPMPA or HDP-(S)-HPMPA were also highly effective (P < 0.001) at preventing mortality in mice infected with VV at 30 mg/kg when treatments were delayed until to 48 or 72 h postinfection, respectively. Protection against both viruses was associated with a significant reduction of virus replication in the liver, spleen, and kidney but not in the lung. These data indicate that HDP-(S)-HPMPA and ODE-(S)-HPMPA are active when given orally against lethal CV and VV infections in mice, and further evaluation is warranted to provide additional information on the potential of these orally active compounds for treatment of human orthopoxvirus infection.


Assuntos
Adenina/análogos & derivados , Antivirais/uso terapêutico , Varíola Bovina/tratamento farmacológico , Organofosfonatos/uso terapêutico , Vaccinia virus/efeitos dos fármacos , Adenina/química , Adenina/farmacocinética , Adenina/uso terapêutico , Administração Oral , Animais , Antivirais/administração & dosagem , Antivirais/farmacocinética , Varíola Bovina/virologia , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/virologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/virologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/virologia , Camundongos , Estrutura Molecular , Organofosfonatos/química , Organofosfonatos/farmacocinética , Baço/efeitos dos fármacos , Baço/metabolismo , Baço/virologia
17.
Antimicrob Agents Chemother ; 51(11): 4118-24, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17724153

RESUMO

The combination of ST-246 and hexadecyloxypropyl-cidofovir or CMX001 was evaluated for synergistic activity in vitro against vaccinia virus and cowpox virus (CV) and in vivo against CV. In cell culture the combination was highly synergistic against both viruses, and the results suggested that combined treatment with these agents might offer superior efficacy in vivo. For animal models, ST-246 was administered orally with or without CMX001 to mice lethally infected with CV. Treatments began 1, 3, or 6 days postinfection using lower dosages than previously used for single-drug treatment. ST-246 was given at 10, 3, or 1 mg/kg of body weight with or without CMX001 at 3, 1, or 0.3 mg/kg to evaluate potential synergistic interactions. Treatment beginning 6 days post-viral inoculation with ST-246 alone only increased the mean day to death at 10 or 3 mg/kg but had no effect on survival. CMX001 alone also had no effect on survival. When the combination of the two drugs was begun 6 days after viral infection using various dosages of the two, a synergistic reduction in mortality was observed. No evidence of increased toxicity was noted with the combination either in vitro or in vivo. These results indicate that combinations of ST-246 and CMX001 are synergistic both in vitro and in vivo and suggest that combination therapy using ST-246 and CMX001 for treatment of orthopoxvirus disease in humans or animals may provide an additional benefit over the use of the two drugs by themselves.


Assuntos
Benzamidas/farmacologia , Citosina/análogos & derivados , Isoindóis/farmacologia , Organofosfonatos/farmacologia , Orthopoxvirus/efeitos dos fármacos , Administração Oral , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Benzamidas/administração & dosagem , Benzamidas/uso terapêutico , Células Cultivadas , Chlorocebus aethiops , Vírus da Varíola Bovina/efeitos dos fármacos , Citosina/farmacologia , Citosina/uso terapêutico , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Humanos , Isoindóis/administração & dosagem , Isoindóis/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Organofosfonatos/uso terapêutico , Infecções por Poxviridae/tratamento farmacológico , Infecções por Poxviridae/virologia , Resultado do Tratamento , Vaccinia virus/efeitos dos fármacos , Células Vero , Replicação Viral/efeitos dos fármacos
18.
Drug Deliv ; 14(3): 119-27, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17454031

RESUMO

Two poly(DL-lactide-co-glycolide) microsphere formulations (A, 10% wt/wt, and B, 23% wt/wt, 1-10 microns) were evaluated for intracellular delivery of rifabutin using the J774 murine and Mono Mac 6 (MM6) human monocytic cell lines. Within 7 days, formulation A released 100% in both cell lines and B released 53 and 67% in the J774 and MM6, respectively. Intracellular release of rifabutin with both formulations caused significant reduction of intracellularly replicating Mycobacterium avium (MAC). In MAC-infected beige mice, formulation B (50 mg, intraperitoneal days 0 and 7) completely eliminated infection by 21 days (p < 0.001), similar to a rifabutin daily oral regimen.


Assuntos
Antibióticos Antituberculose/administração & dosagem , Antibióticos Antituberculose/farmacologia , Macrófagos/microbiologia , Infecção por Mycobacterium avium-intracellulare/tratamento farmacológico , Rifabutina/administração & dosagem , Rifabutina/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Química Farmacêutica , Contagem de Colônia Microbiana , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Microesferas , Monócitos/microbiologia , Infecção por Mycobacterium avium-intracellulare/microbiologia , Mycobacterium phlei/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Teste Bactericida do Soro
19.
Antimicrob Agents Chemother ; 51(2): 689-95, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17116683

RESUMO

ST-246 was evaluated for activity against cowpox virus (CV), vaccinia virus (VV), and ectromelia virus (ECTV) and had an in vitro 50% effective concentration (EC50) of 0.48 microM against CV, 0.05 microM against VV, and 0.07 microM against ECTV. The selectivity indices were >208 and >2,000 for CV and VV, respectively. The in vitro antiviral activity of ST-246 was significantly greater than that of cidofovir, which had an EC50 of 41.1 microM against CV and 29.2 microM against VV, with selectivity indices of >7 and >10, respectively. ST-246 administered once daily by oral gavage to mice infected intranasally with CV beginning 4 h or delayed until 72 h postinoculation was highly effective when given for a 14-day duration using 100, 30, or 10 mg/kg of body weight. When 100 mg/kg of ST-246 was administered to VV-infected mice, a duration of 5 days was sufficient to significantly reduce mortality even when treatment was delayed 24 h postinoculation. Viral replication in liver, spleen, and kidney, but not lung, of CV- or VV-infected mice was reduced by ST-246 compared to levels for vehicle-treated mice. When 100 mg/kg of ST-246 was given once daily to mice infected by the intranasal route with ECTV, treatment for 10 days prevented mortality even when treatment was delayed up to 72 h after viral inoculation. Viral replication in target organs of ECTV-infected mice was also reduced.


Assuntos
Benzamidas/administração & dosagem , Indóis/administração & dosagem , Orthopoxvirus/efeitos dos fármacos , Infecções por Poxviridae/tratamento farmacológico , Administração Oral , Animais , Modelos Animais de Doenças , Feminino , Isoindóis , Camundongos , Especificidade de Órgãos , Orthopoxvirus/fisiologia , Fatores de Tempo , Resultado do Tratamento , Replicação Viral/efeitos dos fármacos
20.
Antiviral Res ; 71(1): 24-30, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16621041

RESUMO

It has been reported previously that some thiosemicarbazone compounds have prophylactic activity against smallpox disease and therapeutic activity against vaccinia virus (VV) infections. In these studies, isatin-beta-thiosemicarbazone (IBT) and marboran were administered once daily by intraperitoneal (ip) injection to mice using 30, 10 or 3 mg/kg for 5 days beginning 24, 48 or 72 h after inoculation with VV or cowpox virus (CV). Both compounds were highly effective (p < 0.01) at preventing mortality due to VV even when treatment was delayed up to 72 h postinfection. In CV-infected mice, neither IBT nor Marboran were effective in preventing mortality at any dosage tested when administered at 24 h postinoculation. Viral replication in liver, spleen and kidney was delayed or reduced by 100-to 10,000-fold by 10 mg/kg of marboran, but not IBT, in VV infections. Neither compound was effective against CV infection. Neither IBT nor marboran treatment of mice cutaneously infected with VV or CV reduced viral replication or clinical disease. These results suggest that this class of compound has little therapeutic potential for orthopoxvirus infections since the in vivo activity against CV, a surrogate virus for variola, is lacking.


Assuntos
Antivirais/farmacologia , Vírus da Varíola Bovina/fisiologia , Hidrazonas/farmacologia , Indóis/farmacologia , Metisazona/farmacologia , Infecções por Poxviridae/tratamento farmacológico , Vaccinia virus/fisiologia , Animais , Anticorpos Antivirais/sangue , Feminino , Camundongos , Camundongos Pelados , Camundongos Endogâmicos BALB C , Infecções por Poxviridae/virologia , Estatísticas não Paramétricas , Análise de Sobrevida , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...