Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Med Chem ; 36(19): 2801-9, 1993 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-8410993

RESUMO

1-Cyclopropyl-6,8-difluoro-1,4-dihydro-7-(2,6-dimethyl-4-pyridinyl)-4-ox o-3-quinolinecarboxylic acid (1), a previously reported potent inhibitor of bacterial DNA gyrase, was found to be interactive with mammalian topoisomerase II (topo II). In a DNA-cleavage assay using topo II isolated from HeLa cells, 1 exhibited an EC50 value of 7.6 microM (VP-16; EC50 = 0.81 microM). A series of analogues modified at the 1-, 2-, 3-, 5-, and 7-positions of 1 were subsequently made and assessed for topo II inhibition. Compound 1 was considerably more potent than derivatives where the 1-substituent was alkyl, aryl, or H, or when N-c-C3H5 was replaced with S. The descarboxyl (i.e., 3-H) analogue had potency comparable to that of 1; when both these compounds were substituted at the 2-position with methyl or phenyl, an interesting relationship between activity and the conformation of the carboxyl group emerged. Upon replacement of the 5-H of 1 with NH2 or F, sustained potency was seen. No enhancement of activity was evident upon replacing the 7-substituent of 1 with other pyridinyl groups, 4-methyl-1-piperazinyl, or pyrrolidinyl groups; however, the 7-(4-hydroxyphenyl) analogue (CP-115,953) was 6-fold more potent than 1. The topo II inhibitory properties of 1 translated to modest in vitro cytotoxicity and in vivo activity versus P388.


Assuntos
Anti-Infecciosos/síntese química , Anti-Infecciosos/farmacologia , Fluoroquinolonas , Quinolonas , Inibidores da Topoisomerase II , Animais , Anti-Infecciosos/química , Células HeLa/efeitos dos fármacos , Humanos , Leucemia P388/tratamento farmacológico , Camundongos , Relação Estrutura-Atividade
2.
J Med Chem ; 38(14): 2541-5, 1995 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-7629793

RESUMO

Supported by the antiherpetic properties of 3-quinolinecarboxamides and the importance of the planar intramolecular H-bonded beta-keto amide pharmacophore, a series of novel conformationally rigid analogues that contain a heterocyclic bridge between the 3- and 4-positions of the quinoline ring have been evaluated. Two isoxazolo-fused derivatives 17 and 23 displayed good in vitro antiherpetic potency that was similar to that of 1, the 3-quinolinecarboxamide that served as the comparison structure for this study. The pyrazolo, pyrrolo, and pyrimido derivatives showed considerably less or no activity. In vitro activity did not translate to in vivo efficacy. For 17, the lack of in vivo activity is likely a consequence of insufficient plasma drug levels (both Cmax and duration) in mice relative to the MIC versus HSV-2.


Assuntos
Antivirais/química , Herpesvirus Humano 2/efeitos dos fármacos , Quinolinas/química , Animais , Antivirais/farmacologia , Modelos Animais de Doenças , Herpes Genital/tratamento farmacológico , Herpesvirus Humano 2/crescimento & desenvolvimento , Espectroscopia de Ressonância Magnética , Camundongos , Quinolinas/farmacologia , Ensaio de Placa Viral
3.
J Med Chem ; 38(8): 1355-71, 1995 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-7731021

RESUMO

Several modifications of the oxazoline ring of WIN 54954, a broad spectrum antipicornavirus compound, have been prepared in order to address the acid lability and metabolic instability of this compound. We have previously shown that the oxadiazole analogue 3 displayed comparable activity against a variety of rhinoviruses and appeared to be stable to acid. A monkey liver microsomal assay was developed to examine the metabolic stability in vitro of both compounds, and it was determined that WIN 54954 displayed 18 metabolic products while 3 was converted to 8 products. Two major products of 3 were determined by LC-MS/MS to be monohydroxylated at each of the terminal methyl groups. Replacement of the methyl on the isoxazole ring with a trifluoromethyl group, while preventing hydroxylation at this position, did not reduce the sensitivity of the molecule to microsomal metabolism at other sites. However, the (trifluoromethyl)oxadiazole 9 not only prevented hydroxylation at this position but also provided protection at the isoxazole end of the molecule, resulting in only two minor products to the extent of 4%. The major product was identified as the monohydroxylated compound 23. The global metabolic protective effect of trifluoromethyl group on the oxadiazole ring was further demonstrated by examining a variety of analogues including heterocyclic replacements of the isoxazole ring. In each case, the trifluoromethyl analogue displayed a protective effect when compared to the corresponding methyl analogue.


Assuntos
Antivirais/farmacologia , Isoxazóis/farmacologia , Microssomos Hepáticos/efeitos dos fármacos , Picornaviridae/efeitos dos fármacos , Animais , Antivirais/química , Antivirais/farmacocinética , Clorofluorcarbonetos de Metano/química , Cromatografia Líquida de Alta Pressão/métodos , Gráficos por Computador , Haplorrinos , Isoxazóis/química , Isoxazóis/farmacocinética , Espectroscopia de Ressonância Magnética , Espectrometria de Massas/métodos , Microssomos Hepáticos/metabolismo , Espectrofotometria Infravermelho
4.
Drug Des Discov ; 15(1): 25-38, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9332829

RESUMO

Novel antiherpetic 3-quinolinecarboxamides were discovered as part of a drug discovery program at Sterling Winthrop Inc. A major goal of this research was to identify novel non-nucleoside agents possessing activity against acyclovir resistant herpes simplex virus. From screening compound libraries in an HSV-2 plaque reduction assay, 1-ethyl-1,4-dihydro-4-oxo-7-(4-pyridinyl)-3-quinolinecarboxamide (1) emerged as an attractive lead structure. By modifying the quinoline ring at the 1-, 2-, 3-, 4-, and 7-positions, analogues were identified that have up to 5-fold increased in vitro potency relative to acyclovir. In a single dose mouse model of infection the 1-(4-FC6H4) analogue 17, one of the most potent derivatives in vitro, displayed comparable oral antiherpetic efficacy to acyclovir at 1/16 the dose; in a multiple dose regimen, however, it was 2-fold less potent. Mechanism of action studies indicate that these new compounds interact with a different, as yet undefined, molecular target than acyclovir.


Assuntos
Antivirais/química , Antivirais/farmacologia , Quinolinas/química , Quinolinas/farmacologia , Aciclovir/farmacologia , Animais , Antivirais/síntese química , Chlorocebus aethiops , Modelos Animais de Doenças , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Herpesvirus Humano 2/efeitos dos fármacos , Humanos , Camundongos , Quinolinas/síntese química , Simplexvirus/efeitos dos fármacos , Relação Estrutura-Atividade , Células Vero , Ensaio de Placa Viral
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa