Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Med Chem ; 33(8): 2127-30, 1990 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-2165160

RESUMO

2,6,8-Trichloro-7-methylpurine (3) was converted to 2-chloro-8,9-dihydro-7-methyl-8-thioxopurin-6(1H)-one (5) by utilizing the difference in reactivity of the 2-, 6-, and 8-positions in the trichloropurine ring system to nucleophilic displacement. Compound 5 was subsequently glycosylated with 1-O-acetyl-2,3,5-tri-O-benzoyl-D-ribofuranose according to the Vorbrüggen procedure to yield 2-chloro-8,9-dihydro-7-methyl-9-(2,3,5-tri-O-benzoyl-beta-D-ribofuranosy l)-8- thioxopurin-6(1H)-one (6). Removal of the benzoyl protecting groups, followed by amination of 7 with liquid ammonia at 150 degrees C, gave 7,8-dihydro-7-methyl-8-thioxoguanosine (2). The structure of compound 2 was confirmed by X-ray crystallographic analysis. Compounds 1 (7,8-dihydro-7-methyl-8-oxoguanosine) and 2 were evaluated for activity in various animal virus infection models. Against banzi, Semliki Forest, and San Angelo viruses in mice, 2 was highly active when administered before virus inoculation.


Assuntos
Antivirais/uso terapêutico , Guanosina/análogos & derivados , Animais , Antivirais/síntese química , Infecções por Bunyaviridae/tratamento farmacológico , Fenômenos Químicos , Química , Feminino , Flavivirus , Guanosina/síntese química , Guanosina/uso terapêutico , Camundongos , Estrutura Molecular , Ratos , Vírus da Floresta de Semliki , Infecções por Togaviridae/tratamento farmacológico , Difração de Raios X
2.
J Med Chem ; 33(10): 2750-5, 1990 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2170645

RESUMO

C-Glycosylation of two guanine analogues, 9-deaza- and 7-deazaguanine, has been achieved under Friedel-Crafts conditions, providing a direct synthetic route to 9-deazaguanosine (4; 2-amino-7-beta-D-ribofuranosyl-5H-pyrrolo[3,2-d]pyrimidin-4(3H)-one) and 8-beta-D-ribofuranosyl-7-deazaguanine (16), respectively. This electrophilic C-glycosylation was applied successfully to six guanine and substituted-guanine analogues resulting in yields of approximately 50%. This represents the first reported C-ribosylation of preformed nitrogen heterocycles isosteric with guanine. These C-nucleosides were evaluated for their ability to provide protection against a lethal Semliki Forest virus infection in mice, relative to 7-thia-8-oxoguanosine which was used as a positive control. Two of the C-nucleosides, 2-amino-6-chloro-5-methyl-7-beta-D-ribofuranosyl-5H-pyrrolo [3,2-d]pyrimidin-4(3H)-one (12) and the corresponding 6-bromo derivative (13), showed good prophylactic activity in this virus model system.


Assuntos
Antivirais/síntese química , Guanosina/análogos & derivados , Animais , Antivirais/química , Antivirais/uso terapêutico , Fenômenos Químicos , Físico-Química , Glicosilação , Guanosina/química , Camundongos , Vírus da Floresta de Semliki , Infecções por Togaviridae/tratamento farmacológico
3.
J Med Chem ; 33(8): 2174-8, 1990 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-2165164

RESUMO

Several guanosine analogues were synthesized in the pyrazolo[3,4-d]pyrimidine ring system with various substituents at the 3-position. The new analogues prepared here include the CH3 (2-amino-3-methyl-1-beta-D-ribofuranosylpyrazolo[3,4-d]pyrimidin-4 (5H)-one, 13a), the phenyl (2-amino-3-phenyl-1-beta-D-ribofuranosylpyrazolo[3,4-d]pyrimidin-4 (5H)-one, 13b), and the NH2 (3,6-diamino-1-beta-D-ribofuranosylpyrazolo[3,4-d]pyrimidin-4(5H)- one, 17) substituted derivatives. These new agents, as well as several other 3-substituted derivatives including H, Br, OCH3, COOH, and oxo, were evaluated for their ability to potentiate certain murine immune functions relative to the known active agent 5-amino-3-beta-D-ribofuranosylthiazolo[4,5-d]pyrimidine-2,7(3H,6H) -dione (4, 7-thia-8-oxoguanosine). The biological evaluation included the (1) ex vivo determination of increased natural killer cell function and (2) in vivo antiviral protection against a lethal challenge of Semliki Forest virus. The 3-unsubstituted (5a) and the 3-bromo (5c) derivatives were found to be the most active immunopotentiators in this series.


Assuntos
Adjuvantes Imunológicos/síntese química , Guanosina/análogos & derivados , Pirazóis/síntese química , Pirimidinonas/síntese química , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/uso terapêutico , Animais , Fenômenos Químicos , Química , Citotoxicidade Imunológica/efeitos dos fármacos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Linfoma/imunologia , Camundongos , Estrutura Molecular , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Pirimidinonas/farmacologia , Pirimidinonas/uso terapêutico , Vírus da Floresta de Semliki , Linfócitos T , Infecções por Togaviridae/prevenção & controle
4.
Antiviral Res ; 26(2): 203-9, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7605116

RESUMO

A novel nucleoside analog, 8-chloro-7-deazaguanosine (8-Cl-7-dzGuo), was evaluated for anti-RNA virus activity in rodents in parallel with the related compound 7-deaza-7-thia-8-oxoguanosine (7-dzTOGuo). Half-daily intraperitoneal (i.p.) doses of each substance administered 24 and 18 h prior to i.p. virus challenge protected the majority of mice infected with banzi, encephalomyocarditis, San Angelo, and Semliki Forest viruses at doses of 25, 50 and 100 mg/kg/day. These compounds at 100 mg/kg/day also protected most suckling rats infected intranasally with rat coronavirus. However, no survival benefit was afforded to treated mice infected intranasally with vesicular stomatitis virus. 8-Cl-7-dzguo was orally active against Semliki Forest virus in mice at 200 and 400 mg/kg/day, whereas 7-dzTOGuo is reported to not be effective orally. In uninfected mice, the two compounds induced similar amounts of interferon following i.p. injections. Interferon was induced by oral treatments with 8-Cl-7-dzGuo but not with 7-dzTOGuo. Fifty percent acute lethal doses to uninfected mice treated i.p. in half-daily doses for one day with 7-deazaguanosine (7-dzGuo), 7-dzTOGuo, and 8-Cl-7-dzGuo were 400, 600 and > 1600 (no mortality at this dose) mg/kg/day, respectively. Daily, i.p. treatments for 14 days with these substances (100 mg/kg/day) showed 7-dzGuo as 100% lethal and the other two substances as not toxic. By virtue of reduced toxicity and oral bioavailability, 8-Cl-7-dzGuo appears to have the greatest clinical potential as an interferon-inducing antiviral agent.


Assuntos
Antivirais/farmacologia , Guanosina/análogos & derivados , Vírus de RNA/efeitos dos fármacos , Viroses/tratamento farmacológico , Animais , Guanosina/farmacologia , Camundongos , Estrutura Molecular , Ratos
5.
Antiviral Res ; 10(6): 253-62, 1988 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-2854956

RESUMO

Ribavirin 5'-sulfamate, a nucleotide analog, inhibited Semliki Forest virus cytopathology by 50% at 10 microM, whereas ribavirin was inactive at less than or equal to 1 mM. Actinomycin D did not reverse (antagonize) the effect of ribavirin 5'-sulfamate against the virus. The compound inhibited amino acid incorporation into macromolecules of uninfected cells but had no appreciable effect on uridine incorporation. Infected cells treated with actinomycin D and nucleotide analog were inhibited in amino acid and uridine incorporation. The compound blocked the formation of the viral RNA polymerase protein in cells, which could account for the inhibited synthesis of new viral RNA. By electrophoresis, inhibition of the synthesis of viral proteins was more pronounced than the inhibition of cellular polypeptides. The analog inhibited the translation of mRNA to protein. Most animals treated intraperitoneally for 7 days with ribavirin 5'-sulfamate at 20 and 40 mg/kg/day starting 2 h before intraperitoneal Semliki Forest virus inoculation survived the otherwise lethal infection.


Assuntos
Antivirais/farmacologia , Ribavirina/farmacologia , Ribonucleosídeos/farmacologia , Vírus da Floresta de Semliki/efeitos dos fármacos , Infecções por Togaviridae/tratamento farmacológico , Animais , Antivirais/uso terapêutico , Divisão Celular/efeitos dos fármacos , Efeito Citopatogênico Viral , RNA Polimerases Dirigidas por DNA/antagonistas & inibidores , Dactinomicina/farmacologia , Feminino , Camundongos , Estrutura Molecular , Biossíntese de Proteínas/efeitos dos fármacos , Puromicina/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/efeitos dos fármacos , RNA Viral/biossíntese , RNA Viral/efeitos dos fármacos , Ribavirina/análogos & derivados , Ribavirina/uso terapêutico , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/crescimento & desenvolvimento , Células Vero
6.
Antiviral Res ; 13(2): 91-102, 1990 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-1693058

RESUMO

7-Thia-8-oxoguanosine is a novel biological response modifier with broad-spectrum antiviral activity against many DNA and RNA viruses in vivo. Since two of its properties are to induce interferon and to activate natural killer (NK) cells, we investigated the roles of the lymphokine and NK cells in the antiviral activity of the compound against Semliki Forest virus. Antibody to interferon alpha/beta could completely abolish the protective activity of the nucleoside against virus infection in mice, whereas antibodies to interferons beta and gamma could not, indicating that interferon alpha was of major importance to confer protection to the animals. Reduced activation of NK cells was also observed in mice treated with 7-thia-8-oxoguanosine and antibody to interferon alpha/beta. The role of NK cells in the protective activity of the compound was directly assessed in beige mice or in Swiss Webster mice treated with asialo GM1 antibody. In both experiments, the animals were protected from lethal virus infection by treatment with nucleoside. Spleen cells primed by 7-thia-8-oxoguanosine and adoptively transferred to untreated mice could not save them from virus-induced mortality. These three results provide evidence that natural killer cells activated by 7-thia-8-oxoguanosine play a minimal role in protection from acute Semliki Forest virus infections in mice.


Assuntos
Citotoxicidade Imunológica , Guanosina/análogos & derivados , Interferons/fisiologia , Células Matadoras Naturais/imunologia , Vírus da Floresta de Semliki/efeitos dos fármacos , Infecções por Togaviridae/tratamento farmacológico , Animais , Anticorpos Antivirais/imunologia , Células Cultivadas , Quimioterapia Combinada , Feminino , Guanosina/farmacologia , Dose Letal Mediana , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Floresta de Semliki/imunologia , Infecções por Togaviridae/imunologia
7.
Antiviral Res ; 13(1): 1-10, 1990 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-2159260

RESUMO

The heterocycle, 4,6-dibenzamidopyrazolo[3,4-d]pyrimidine (DBAPP), inhibited cytopathology induced by human, mouse, and vervet monkey cytomegaloviruses (CMV) in vitro at 0.2 to 0.5 microM, but did not inhibit cell replication at less than or equal to 30 microM. Herpes simplex viruses were unaffected by the inhibitor. The antiviral agent ganciclovir was effective against these CMVs at 3-10 microM in parallel assays. DBAPP and ganciclovir were synergistic inhibitors when used in combination. The heterocycle was only active if applied to cells before virus replication, indicating that it inhibited virus adsorption. Cells pre-treated 1 h with 30 microM DBAPP, then extensively rinsed, were resistant to infection by mouse CMV even 3 days after removal of the inhibitor. Human and monkey CMVs were able to infect cells and replicate within 24 h of drug removal. When virus and DBAPP were combined together then dialyzed to remove the compound, mouse CMV infectivity was decreased 1.7 logs, whereas human CMV and monkey CMV infectivity titers were relatively unaffected. Treatment of mice with DBAPP twice a day for 7 days starting 6 h after mouse CMV inoculation caused a moderate increase in number of survivors at 30 mg/kg. Cell to cell spread of the virus may account for poor efficacy of the compound when added after virus infection. DBAPP may serve as a tool to explore aspects of CMV adsorption or to characterize the cellular component of the CMV receptor.


Assuntos
Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Adsorção , Animais , Células Cultivadas , Cercopithecus , Citomegalovirus/fisiologia , Efeito Citopatogênico Viral/efeitos dos fármacos , Depressão Química , Sinergismo Farmacológico , Ganciclovir/farmacologia , Humanos , Camundongos , Simplexvirus/efeitos dos fármacos
8.
Antiviral Res ; 10(6): 263-77, 1988 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-2854957

RESUMO

Eight sugar-modified pyrrolopyrimidine nucleoside analogs related to the antibiotic sangivamycin were evaluated in cell culture against herpes simplex types 1 (HSV-1) and 2 (HSV-2), cytomegalovirus (CMV), adenovirus, and visna virus. Five of the compounds were highly active against most of the viruses with 50% inhibition (ED50) values of 1-10 microM. The selectivity of the agents was low, with inhibition of uninfected cell proliferation occurring within 5-fold that of the virus ED50 for most of the viruses. The compounds did not possess RNA virus-inhibitory activity when evaluated against certain myxo-, paramyxo-, picorna-, reo-, rhabdo-, and togaviruses. Two of the nucleosides were tested further in a cell line persistently infected with Friend leukemia virus where they were inhibitory to both virus yield and cell proliferation at 4-5 microM. Several of the sangivamycin analogs were tested in animal models using a twice-a-day treatment regimen. They proved to be inactive against HSV-1, murine CMV and/or Friend leukemia virus infections in mice.


Assuntos
Antivirais/farmacologia , Vírus de DNA/efeitos dos fármacos , Retroviridae/efeitos dos fármacos , Adenoviridae/efeitos dos fármacos , Animais , Antivirais/uso terapêutico , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Citomegalovirus/efeitos dos fármacos , Infecções por Citomegalovirus/tratamento farmacológico , Vírus da Leucemia Murina de Friend/efeitos dos fármacos , Herpes Simples/tratamento farmacológico , Humanos , Leucemia Experimental/tratamento farmacológico , Camundongos , Estrutura Molecular , Nucleosídeos de Pirimidina/farmacologia , Nucleosídeos de Pirimidina/uso terapêutico , Simplexvirus/efeitos dos fármacos , Células Tumorais Cultivadas , Células Vero , Vírus Visna-Maedi/efeitos dos fármacos
9.
Antiviral Res ; 6(6): 343-53, 1986 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-3022644

RESUMO

The inhibitory effects of selenazofurin and ribavirin on influenza A and B virus infections in mice were compared. Both compounds, when administered intraperitoneally (i.p.), reduced lung consolidation and prolonged mean day of death, but ribavirin more effectively increased survivor number and lowered lung viral hemagglutinin (HA) titers. Lung HA titers often increased in selenazofurin-treated animals. To determine the most appropriate i.p. treatment schedule, influenza A virus-infected mice were treated once, twice or thrice daily for 7-9 days, or once only. Treatment once daily for 9 days beginning 4 h pre-virus exposure, for 3 days beginning 24 h post-virus exposure, or once only 48 h post-virus exposure was most effective. Body temperature, which usually declined during infection, increased to near-normal levels in animals treated with selenazofurin, especially in animals treated a single time or for 3 days with high dose levels. Selenazofurin was well tolerated at a dose of 50 mg/kg administered twice daily, and at 400 mg/kg administered once only. Rectal temperatures temporarily declined following every other day treatment with 400 mg/kg.


Assuntos
Compostos Organosselênicos , Infecções por Orthomyxoviridae/tratamento farmacológico , Ribavirina/uso terapêutico , Ribonucleosídeos/uso terapêutico , Selênio/uso terapêutico , Animais , Avaliação Pré-Clínica de Medicamentos , Feminino , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Injeções Intraperitoneais , Pulmão/microbiologia , Camundongos , Ribavirina/administração & dosagem , Ribonucleosídeos/administração & dosagem , Selênio/administração & dosagem
10.
Arch Virol ; 107(3-4): 253-60, 1989.
Artigo em Inglês | MEDLINE | ID: mdl-2554856

RESUMO

Cell culture systems were developed for rapid antiviral drug screening, using murine cytomegalovirus (MCMV) as an alternative to the slower growing human CMV. Since previous assay methods with MCMV employed mouse embryo fibroblasts (MEF cells), which are labor intensive to prepare and die off after 3-4 passages from primary culture, identification of virus-susceptible continuous cell lines was desirable. Three cell lines were found useful for assaying MCMV: C127I, SC-1, and 3T3. The antiviral agents acyclovir, ganciclovir, 5-fluoroarabinofuranosylcytosine, and 2'-fluoro-2'-deoxy-5-iodoarabinofuranosylcytosine were evaluated in the 3 continuous cell lines and in MEF cells. The 50% virus- or cell-inhibitory concentration values determined for each compound did not vary much from cell to cell. MEF cells were 10-fold more sensitive than the other cell lines to quantify virus from mouse organs, however. Virus propagated in 3T3 and SC-1 cells were as virulent to mice as salivary gland virus, whereas virus from MEF and C127I cells was more attenuated. Overall, C127I cells were judged to be the best for large scale antiviral screening in vitro, but MEF was the cell type of choice for titration of viruses from mouse organs and tissues.


Assuntos
Antivirais/farmacologia , Linhagem Celular , Citomegalovirus/crescimento & desenvolvimento , Cultura de Vírus , Animais , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/patogenicidade , Efeito Citopatogênico Viral , Avaliação Pré-Clínica de Medicamentos , Feminino , Camundongos , Ensaio de Placa Viral , Virulência , Replicação Viral/efeitos dos fármacos
11.
Chemotherapy ; 33(2): 141-50, 1987.
Artigo em Inglês | MEDLINE | ID: mdl-3032526

RESUMO

Vidarabine (9-beta-D-arabinofuranosyladenine) prepared in a 70% dimethyl sulfoxide vehicle was applied topically to type 1 herpesvirus-induced cutaneous lesions on guinea pigs and athymic nude mice. Treatments were 3 or 5 times daily for 7 days beginning 24 h after virus exposure. Against infections in guinea pigs induced by a thymidine kinase-positive virus strain, either treatment schedule effectively inhibited mean lesion score, lesion size, appearance of new lesions, and reduced lesion virus titers. Therapy was similarly effective against infections in guinea pigs induced by a thymidine kinase-negative virus strain, except that lesion virus titers were somewhat increased in animals treated 3 times daily. Treatment 5 times daily was most efficacious against both virus strains. Treatment 3 times daily of mice infected with a thymidine kinase-negative virus was not effective, but treatment 5 times daily significantly inhibited lesion score and size and reduced lesion virus titer by 37%. Toxicity controls exhibited no signs of skin irritation, although guinea pigs treated 5 times daily experienced some transient weight loss.


Assuntos
Infecções por Herpesviridae/tratamento farmacológico , Dermatopatias Infecciosas/tratamento farmacológico , Vidarabina/uso terapêutico , Animais , Dimetil Sulfóxido , Feminino , Cobaias , Infecções por Herpesviridae/microbiologia , Camundongos , Camundongos Nus , Pele/patologia , Dermatopatias Infecciosas/microbiologia , Dermatopatias Infecciosas/patologia , Especificidade da Espécie , Timidina Quinase/antagonistas & inibidores , Vidarabina/administração & dosagem
12.
J Biol Response Mod ; 9(1): 24-32, 1990 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-2156954

RESUMO

A novel thiazolopyrimidine nucleoside, 5-amino-3-beta-D-ribofuranosylthiazolo[4,5-d]pyrimidine-2,7(3H,6H) -dione (7-thia-8-oxoguanosine), was evaluated for antiviral activity in rodent models, and at 50-200 mg/kg prevented death in mice inoculated intraperitoneally (i.p.) with Semliki Forest, San Angelo, and banzi viruses when administered i.p. before virus challenge. Similarly, the nucleoside was effective against an intranasal challenge of rat coronavirus in suckling rats, with activity present when treatment started as late as 4 h after virus inoculation. Protection was observed against herpes type 1 and murine cytomegalovirus (both inoculated i.p.) infections, and encephalitis induced by intracerebral inoculation of a human coronavirus in mice. Friend leukemia virus splenomegaly was more severe in drug-treated animals than in placebos. This immune modulator is promising for the treatment of animal and human diseases.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Antivirais , Guanosina/análogos & derivados , Animais , Antivirais/uso terapêutico , Modelos Animais de Doenças , Feminino , Guanosina/uso terapêutico , Camundongos , Gravidez , Ratos , Ratos Endogâmicos F344 , Vírus da Floresta de Semliki , Infecções por Togaviridae/tratamento farmacológico , Viroses/tratamento farmacológico
13.
Antimicrob Agents Chemother ; 28(3): 375-7, 1985 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-4073860

RESUMO

Activity of the new antiviral compound selenazofurin was compared with the known active compounds ribavirin and amantadine against influenza A and B viruses. In experiments with Madin Darby canine kidney cells, selenazofurin inhibited the cytopathic effect and yield of influenza A/NWS/33 virus, with 50% effective dose ranges of 0.7 to 1.4 micrograms/ml (virus rating [VR], 1.3 to 1.4). The 50% effective dose range for ribavirin was 1.2 to 1.6 micrograms/ml (VR, 1.0 to 1.3), and for amantadine it was 9 micrograms/ml (VR, 0.9). Selenazofurin and ribavirin were similarly inhibitory to influenza B/Lee/40 virus, whereas amantadine was inactive. Selenazofurin appeared somewhat cytotoxic in these studies at concentrations as low as 1 micrograms/ml.


Assuntos
Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Compostos Organosselênicos , Ribonucleosídeos/farmacologia , Selênio/farmacologia , Amantadina/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Efeito Citopatogênico Viral/efeitos dos fármacos , Cães , Técnicas In Vitro , Ribavirina/farmacologia , Replicação Viral/efeitos dos fármacos
14.
Antimicrob Agents Chemother ; 33(9): 1487-92, 1989 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-2817849

RESUMO

A novel immunopotentiating agent, 5-amino-3-beta-D-ribofuranosylthiazolo [4,5-d]pyrimidine-2,7(3H,6H)-dione (7-thia-8-oxoguanosine), lacks virus-inhibitory properties in vitro but induces interferon and potentiates immune functions, such as natural killer cell activity. It was evaluated in rodent models to determine the spectrum of antiviral activity and effective treatment regimens. At 50 to 200 mg/kg given as single or divided intraperitoneal (i.p.) doses 1 day before virus inoculation, significant protection was afforded to mice infected i.p. with Semliki Forest, San Angelo, banzi, and encephalomyocarditis viruses. Similarly, suckling rats were protected from an intranasal challenge with rat coronavirus. Against San Angelo virus, treatments could be delayed to 1 day post-virus inoculation and still show a beneficial effect. The compound was moderately effective in mice infected i.p. with herpes simplex virus type 2 or intranasally with vesicular stomatitis virus. No activity was seen against influenza B virus in mice when the analog was administered one time pre-virus inoculation or in multiple doses given before and after the virus inoculation. Nor was there a prophylactic effect against herpetic skin lesions on mice. This immune modulator may have promise for the treatment of a variety of virus infections.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antivirais , Adjuvantes Imunológicos/uso terapêutico , Animais , Animais Lactentes , Feminino , Guanosina/farmacologia , Indutores de Interferon , Células Matadoras Naturais/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Pelados , Gravidez , Ratos , Viroses/tratamento farmacológico , Viroses/microbiologia
15.
Antimicrob Agents Chemother ; 35(1): 152-7, 1991 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1707603

RESUMO

The nucleotide analog 7-deazaguanosine has not previously been reported to possess biological (antiviral or antitumor) properties in cell culture or in vivo. Up to 10(5) U of interferon per ml was detected in mouse sera 1 to 4 h following oral (200-mg/kg of body weight) and intraperitoneal (50-mg/kg) doses of the compound. 7-Deazaguanosine also caused significant activation of natural killer and phagocytic cells but did not augment T- and B-cell blastogenesis. Intraperitoneal treatments of 50, 100, and 200 mg/kg/day administered 24 and 18 h before virus inoculation were highly protective in mice inoculated with lethal doses of Semliki Forest or San Angelo viruses. Less but still significant survivor increases were evident in treated mice infected with banzi or encephalomyocarditis viruses. In most cases, the degree of antiviral activity was similar to that exhibited by the biological response modifier 7-thia-8-oxoguanosine. 7-Thia-8-oxoguanosine was more potent than 7-deazaguanosine against encephalomyocarditis virus in mice, however. Oral efficacy was achieved with 7-deazaguanosine treatments of greater than or equal to 100 mg/kg against all virus infections, whereas 7-thia-8-oxoguanosine is reported to be devoid of oral activity in rodents. Thus, 7-deazaguanosine represents the first reported orally active nucleoside biological response modifier exhibiting broad-spectrum antiviral activity against particular types of RNA viruses.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antivirais/farmacologia , Guanosina/análogos & derivados , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/toxicidade , Administração Oral , Animais , Antivirais/administração & dosagem , Antivirais/toxicidade , Feminino , Guanosina/administração & dosagem , Guanosina/farmacologia , Guanosina/toxicidade , Injeções Intraperitoneais , Interferons/biossíntese , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Camundongos , Viroses/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA