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










Base de dados
Intervalo de ano de publicação
1.
Indian J Pharm Sci ; 72(6): 778-81, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21969752

RESUMO

A pyrimidne nucleobase, 5-phenylthio-2,4-bisbenzyloxypyrimidine and its analogs were synthesized and scanned for in vitro antifungal activity using cup-plate and macrobroth dilution method against Candida albicans, Aspergillus niger, Aspergillus flavus and Aspergllus fumigatus. In the cup-plate method, 5-phenylthio-2,4-bisbenzyloxypyrimidine showed very good antifungal activity compared to clotrimazole at the concentrations of 100 and 1000 µg/ml and in the macrobroth dilution method, it showed comparable activity with respect to standard drugs fluconazole and itraconaole. In vivo antifungal activity of 5-phenylthio-2,4-bisbenzyloxypyrimidine at the dose levels of 10 and 30 mg/kg was carried by causing systemic infection of mice using the same fungi used in in vitro testing. The results from in vivo studies with 5-phenylthio-2,4-bisbenzyloxypyrimidine and fluconazole indicated that 5-phenylthio-2,4-bisbenzyloxypyrimidine had similar potency as fluconazole at both dose levels.

2.
Indian J Pharm Sci ; 71(6): 672-7, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20376222

RESUMO

4-Substituted-5-iodo-2-benzylthiopyrimidines were prepared efficiently in three steps. 2-Benzylthiopyrimidine on iodination in presence of base gave 5-iodo-2-benzylthiopyrimidine (1), which on chlorination with excess of POCl(3) furnished 4-chloro-5-iodo-2-benzylthiopyrimidine (2). Reaction of 2 with substituted aromatic amines, 2-aminopyridine and hydrazine hydrate yielded 4-amino-5-iodo-2-benzylthiopyrimidines 3(a-e), (3f) and (3g) respectively. Further, 4-hydrazino-5-iodo-2-benzylthiopyrimidine on condensation with substituted aromatic and heterocyclic aldehydes afforded the corresponding schiff bases 4(a-h). The structure of synthesized compounds have been established by spectral studies and elemental analysis. Synthesized compounds have been screened for antimicrobial activity. Compound 3f exhibited good antifungal activity against A. niger. The compounds 4a, 4c, 4d, 4g and 4h exhibited good antibacterial activity.

3.
Cancer Chemother Pharmacol ; 48(2): 145-50, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11565561

RESUMO

PURPOSE: The purpose of this investigation was to evaluate the ability of oral PTAU, 5-(phenylthio)acyclouridine, to increase the concentration of endogenous plasma uridine. PTAU is a new potent and specific inhibitor of uridine phosphorylase (UrdPase, EC 2.4.2.3), the enzyme responsible for uridine catabolism. This compound was designed as a lipophilic inhibitor in order to facilitate its access to the liver and intestine, the main organs involved in uridine catabolism. METHODS: PTAU was administered to mice orally and parenterally. The plasma levels of PTAU as well as those of uridine and its catabolite uracil were measured by HPLC, and pharmacokinetic analysis was performed. RESULTS: PTAU was fully adsorbed after oral administration (over 100% oral bioavailability) and no PTAU metabolites were detected. PTAU administered orally had no apparent toxicity at doses up to 120 mg/kg per day for 5 days. Parenteral administration of PTAU at 30, 45 and 60 mg/kg increased the concentration of endogenous plasma uridine (1.8 +/- 0.2 microM) by approximately six-, seven-, and nine-fold, respectively. Plasma uridine concentration remained higher than control values until 8 h after PTAU administration. Similar results were obtained following oral administration of PTAU. The baseline concentrations of endogenous plasma uridine were increased by approximately six-, seven- and ten-fold by oral administration of PTAU at 30, 45 and 60 mg/kg, respectively, and remained higher than the controls until 8 h after PTAU administration. PTAU did not alter the concentration of endogenous plasma uracil. CONCLUSION: The effectiveness of the PTAU in elevating and sustaining high plasma uridine concentrations may be useful in rescuing or protecting the host from toxicities of various chemotherapeutic pyrimidine analogues as well as in the management of medical disorders that respond to the administration of uridine.


Assuntos
Inibidores Enzimáticos/farmacologia , Tiouracila/análogos & derivados , Tiouracila/farmacologia , Uridina/sangue , Animais , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/toxicidade , Feminino , Camundongos , Tiouracila/sangue , Tiouracila/farmacocinética , Tiouracila/toxicidade , Uridina Fosforilase/antagonistas & inibidores
4.
Cancer Chemother Pharmacol ; 46(3): 235-40, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11021741

RESUMO

PURPOSE: The purpose of this investigation was to study the effects of combining oral 5-(phenylselenenyl)acyclouridine (PSAU) with 2',3',5'-tri-O-acetyluridine (TAU) on the levels of plasma uridine in mice. PSAU is a new lipophilic and potent inhibitor of uridine phosphorylase (UrdPase, EC 2.4.2.3), the enzyme responsible for uridine catabolism. PSAU has 100% oral bioavailability and is a powerful enhancer of the bioavailability of oral uridine. TAU is a prodrug of uridine and a far superior source of uridine than uridine itself. METHODS: Oral TAU was administered to mice alone or with PSAU. The plasma levels of uridine and its catabolites as well as PSAU were measured using HPLC and pharmacokinetic analysis was performed. RESULTS: Oral administration of 2000 mg/kg TAU increased plasma uridine by over 250-fold with an area under the curve (AUC) of 754 micromol x h/l. Coadministration of PSAU at 30 and 120 mg/kg with TAU further improved the bioavailability of plasma uridine resulting from the administration of TAU alone by 1.7- and 3.9-fold, respectively, and reduced the Cmax and AUC of plasma uracil. CONCLUSION: The exceptional effectiveness of PSAU plus TAU in elevating and sustaining a high plasma uridine concentration could be useful in the management of medical disorders that are remedied by administration of uridine, as well as the rescue or protection from host toxicities of various chemotherapeutic pyrimidine analogues.


Assuntos
Inibidores Enzimáticos/farmacologia , Compostos Organosselênicos/farmacologia , Pró-Fármacos/farmacologia , Uracila/análogos & derivados , Uracila/farmacologia , Uridina/sangue , Uridina/farmacologia , Acetatos , Animais , Disponibilidade Biológica , Sinergismo Farmacológico , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/farmacocinética , Feminino , Camundongos , Compostos Organosselênicos/sangue , Compostos Organosselênicos/farmacocinética , Pró-Fármacos/farmacocinética , Uracila/sangue , Uracila/farmacocinética , Uridina/análogos & derivados , Uridina/farmacocinética , Uridina Fosforilase/antagonistas & inibidores
5.
Biochem Pharmacol ; 60(5): 687-92, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10927027

RESUMO

The effect of co-administration of 5-(phenylselenenyl)acyclouridine (PSAU), a new uridine phosphorylase (UrdPase, EC 2.4.2.3) inhibitor, on the efficacy of 5-fluoro-2'-deoxyuridine (FdUrd) was tested against murine colon C26-10 tumor xenografts. In contrast to our previous results with human tumors, co-administration of PSAU with FdUrd decreased instead of increasing the efficacy of FdUrd against tumor growth. However, co-administration of PSAU with FdUrd (300 mg/kg/day) protected the mice completely from the 83% mortality induced by the same dose of FdUrd alone. Enzyme studies indicated that UrdPase in colon C26-10 tumors is responsible for the catabolism of FdUrd to 5-fluorouracil (FUra), as colon C26-10 tumors do not have thymidine phosphorylase (dThdPase, EC 2.4.2.4). In contrast, colon C26-10 tumors had extraordinarily high UrdPase activity (300 micromol/min/mg protein), which was at least 200-fold higher than the highest UrdPase activity in any of the human xenografts we tested previously. Furthermore, the activities of UrdPase and orotate phosphoribosyltransferase (OPRTase, EC 2.4.2.10) were 192- and 2-fold higher, respectively, while that of dihydrouracil dehydrogenase (EC 1.3.1.2) was 1000-fold lower in the tumor than in the host liver. It is suggested that FdUrd exerts its anticancer effects against colon C26-10 tumors mainly through the catabolism of FdUrd to FUra by UrdPase, which then could be anabolized to 5-fluorouridine 5'-monophosphate (FUMP) by OPRTase and ultimately to other toxic 5-fluorouridine nucleotides, hence inducing the observed FdUrd toxic effects. Co-administration of PSAU with FdUrd inhibited UrdPase and the catabolism of FdUrd to FUra. This would result in the observed reduction of the antitumor efficacy of FdUrd. In addition, the increase in plasma uridine concentration induced by PSAU as well as the catabolism of FUra by the high dihydrouracil dehydrogenase activity in the liver also may have circumvented any residual FUra toxic effects against the host. These results clearly demonstrate that the anticancer efficacy of the combination of UrdPase inhibitors and FdUrd is not general and is dependent largely on the type of tumor under treatment and the mode of FdUrd metabolism in these tumors.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Floxuridina/farmacologia , Compostos Organosselênicos/farmacologia , Uracila/análogos & derivados , Uracila/farmacologia , Uridina Fosforilase/antagonistas & inibidores , Animais , Interações Medicamentosas , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Transplante Heterólogo , Células Tumorais Cultivadas
6.
Biochem Pharmacol ; 60(6): 851-6, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10930540

RESUMO

5-Phenylthioacyclouridine (PTAU or 1-[(2-hydroxyethoxy)methyl]-5-phenylthiouracil) was synthesized as a highly specific and potent inhibitor of uridine phosphorylase (UrdPase, EC 2.4.2.3). PTAU has inhibition constant (K(is)) values of 248 and 353 nM towards UrdPase from mouse and human livers, respectively. PTAU was neither an inhibitor nor a substrate for thymidine phosphorylase (EC 2.4.2.4), uridine-cytidine kinase (EC 2. 7.1.48), thymidine kinase (EC 2.7.1.21), dihydrouracil dehydrogenase (EC 1.3.1.2), orotate phosphoribosyltransferase (EC 2.4.2.10), or orotidine 5'-monophosphate decarboxylase (EC 4.1.2.23), the enzymes that could utilize the substrate (uridine or thymidine) or products (uracil or thymine) of UrdPase. Different isomers of 5-tolylthiouracil also were synthesized and tested as inhibitors of UrdPase. The meta-substituted isomer was 3- to 4-fold more potent as an inhibitor of UrdPase than the para- or ortho-substituted isomers. These data indicate that the hydrophobic pocket in the active site of UrdPase adjacent to the 5-position of the pyrimidine ring can accommodate the meta-substituted 5-phenyluracils better than the other isomers, leading to improved inhibition. Therefore, it is anticipated that the potency of PTAU can be increased further by the addition of certain hydrophobic groups at the meta position of the phenyl ring. PTAU has potential usefulness in the therapy of cancer and AIDS as well as other pathological and physiological disorders that can be remedied by the administration of uridine.


Assuntos
Inibidores Enzimáticos/farmacologia , Tiouracila/farmacologia , Uridina Fosforilase/antagonistas & inibidores , Animais , Ligação Competitiva , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Feminino , Humanos , Cinética , Fígado/enzimologia , Camundongos , Tiouracila/análogos & derivados , Tiouracila/síntese química , Tiouracila/química , Tiouracila/farmacocinética
7.
Cancer Chemother Pharmacol ; 45(5): 351-61, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10803917

RESUMO

PURPOSE: The purpose of this investigation was to evaluate the efficacy of oral 5-(phenylselenenyl)-acyclouridine (PSAU) in increasing endogenous plasma uridine concentration as well as its ability to improve the bioavailability of oral uridine. PSAU is a new potent and specific inhibitor of uridine phosphorylase (Urd-Pase, EC 2.4.2.3), the enzyme responsible for uridine catabolism. This compound was designed as a lipophilic inhibitor in order to facilitate its access to the liver and intestine, the main organs involved in uridine catabolism. METHODS: Oral PSAU was administered orally to mice alone or with uridine. The plasma levels of PSAU as well as uridine and its catabolites were measured using high-performance liquid chromatography and pharmacokinetic analysis was performed. RESULTS: PSAU has an oral bioavailability of 100% and no PSAU metabolites were detected. PSAU has no apparent toxicity at high doses. Oral administration of PSAU at 30 and 120 mg/kg increased baseline concentration of endogenous plasma uridine (2.6 +/- 0.7 microM) by 3.2- and 8.7-fold, respectively, and remained three- and six-fold higher, respectively, than the controls for over 8 h. PSAU, however, did not alter the concentration of endogenous plasma uracil. Co-administration of PSAU with uridine elevated the concentration of plasma uridine over that resulting from the administration of either alone, and reduced the peak plasma concentration (C(max)) and area under the curve (AUC) of plasma uracil. Co-administration of PSAU at 30 mg/kg and 120 mg/kg improved the low bioavailability of oral uridine (7.7%) administered at 1,320 mg/kg by 4.8- and 4.2-fold, respectively, and reduced the AUC of plasma uracil from 1,421 to 787 micromol/h x l and 273 micromol/h x l, respectively. Similar results were observed when PSAU was co-administered with lower doses of uridine. Oral PSAU at 30 mg/kg and 120 mg/kg improved the bioavailability of oral 330 mg/kg uridine by 5.2- and 8.9-fold, and that of oral 660 mg/kg uridine by 6.4- and 9.0-fold, respectively. However, the reduction in the AUC values of plasma uracil was less dramatic than that seen when the high dose of 1,320 mg/kg uridine was used. CONCLUSION: The effectiveness of the PSAU plus uridine combination in elevating and sustaining high plasma uridine concentration may be useful to rescue or protect from host toxicity of various chemotherapeutic pyrimidine analogs as well as in the management of medical disorders that are remedied by administration of uridine.


Assuntos
Inibidores Enzimáticos/farmacologia , Compostos Organosselênicos/farmacologia , Uracila/análogos & derivados , Uridina Fosforilase/antagonistas & inibidores , Uridina/sangue , Animais , Disponibilidade Biológica , Feminino , Camundongos , Compostos Organosselênicos/farmacocinética , Uracila/farmacocinética , Uracila/farmacologia
8.
Biochem Pharmacol ; 51(12): 1687-700, 1996 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-8687484

RESUMO

One hundred and fifty analogues of uridine, with various modifications to the uracil and pentose moieties, have been tested and compared with uridine with respect to their potency to bind to uridine phosphorylase (UrdPase, EC 2.4.2.3) from Toxoplasma gondii. The effects of the alpha- and beta-anomers, the L- and D-enantiomers, as well as restricted syn and anti rotamers, on binding were examined. Pseudo-, lyxo-, 2,3'-anhydro-2'-deoxy-, 6,5'-cyclo-, 6,3'-methano-, O5',6-methano- and carbocyclic uridines did not bind to the enzyme. Ribosides bound better than the corresponding xylosides, which were better than the deoxyribosides. The binding of deoxyribosides was in the following manner: 2',3'-dideoxynucleosides > 2',5'-dideoxynucleosides > 2'-deoxyribosides > 3'- and 5'-deoxyribosides. alpha-2'-Deoxyribosides bound to the enzyme, albeit less tightly than the corresponding beta-anomers. The acyclo- and 2,2'-anhydrouridines bound strongly, with the 2,2'-anhydro-derivatives being the better ligands. 2,5'-Anhydrouridine bound to UrdPase less effectively than 2,2'-anhydrouridine and acyclouridine. Arabinosyluracil was at best a very poor ligand, but bound better if a benzyl group was present at the 5-position of the pyrimidine ring. This binding was enhanced further by adding a 5-benzyloxybenzyl group. A similar enhancement of the binding by increased hydrophobicity at the 5-position of the pyrimidine ring was observed with ribosides, alpha- and beta-anomers of the 2'-deoxyribosides, acyclonucleosides, and 2,2'-anhydronucleosides. Among all the compounds tested, 5-(benzyloxybenzyl)-2,2'-anhydrouridine was identified as the best ligand of T. gondii UrdPase with an apparent Ki value of 60 +/- 3 nM. It is concluded that the presence of an N-glycosyl bond is a prerequisite for a nucleoside ligand to bind to T. gondii UrdPase. On the other hand, the presence of a 2'-, 3'-, or 5'-hydroxyl group, or an N-glycosyl bond in the beta-configuration, enhanced but was not essential for binding. Furthermore, the potency of the binding of 2,2'-anhydrouridines (fixed high syn isomers) in contrast to the weaker binding of the 6,1'-anhydro- or 2,5'-anhydrouridines (fixed syn isomers), and the complete lack of binding of the 6,5'-cyclo, O5',6-methano- and 6,3'-methanouridines (fixed anti isomers) to T. gondii UrdPase indicate that the binding of ligands to this enzyme is in the syn/high syn conformation around the N-glycosyl bond. The results also indicate that the parasite but not the mammalian host UrdPase can participate in hydrogen bonding with N3 of the pyrimidine ring of nucleoside ligands. T. gondii UrdPase also has a larger hydrophobic pocket adjacent to the C5 of the pyrimidine moiety than the host enzyme, and can accommodate modifications in the pentose moiety which cannot be tolerated by the host enzyme. Most prominent among these modifications is the absence and/or lack of the ribo orientation of the 3'-hydroxyl group, which is a requirement for a ligand to bind to mammalian UrdPase. These differences between the parasite and host, enzymes can be useful in designing specific inhibitors or "subversive" substrates for T. gondii UrdPase.


Assuntos
Relação Estrutura-Atividade , Uridina Fosforilase/efeitos dos fármacos , Uridina Fosforilase/metabolismo , Uridina/análogos & derivados , Uridina/química , Animais , Feminino , Ligantes , Camundongos , Camundongos Endogâmicos , Toxoplasma
9.
Antimicrob Agents Chemother ; 38(10): 2409-14, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7840579

RESUMO

Acyclic 6-phenylselenenyl- and 6-phenylthiopyrimidine derivatives are potent and specific inhibitors of human immunodeficiency virus type 1 (HIV-1). The development of in vitro resistance to two derivatives, 5-ethyl-1-(ethoxymethyl)-(6-phenylthio)-uracil (E-EPU), was evaluated by serial passage of HIV-1 in increasing concentrations of inhibitor. HIV-1 variants exhibiting > 500-fold resistance to E-EPSeU and E-EPU were isolated after sequential passage in 1, 5, and 10 microM inhibitor. The resistant variants exhibited coresistance to related acyclic 6-substituted pyrimidines and the HIV-1-specific inhibitors (+)-(5S)-4,5,6,7-tetrahydro-5- pyrimidines and the HIV-1-specific inhibitors (+)-(5S)-4,5,6,7-tetrahydro-5- methyl-6-(3-methyl-2-butenyl)imidazo[4,5,1-jk]benzodiazepin-2(1H)- thione (TIBO R82150) and nevirapine, but remained susceptible to 3'-azido-3'-deoxythymidine, 2',3'-dideoxycytidine, 2',3'-dideoxyinosine, and phosphonoformic acid. DNA sequence analysis of reverse transcriptase (RT) derived from E-EPSeU-resistant virus identified a Tyr (TAT)-to-Cys (TGT) mutation at either codon 188 (Cys-188; 9 of 15 clones) or codon 181 (Cys-181; 5 of 15 clones). The same amino acid changes were found in RT from E-EPU-resistant virus, but the Cys-181 mutation was more common (9 of 10 clones) than the Cys-188 mutation (1 of 10 clones). Site-specific mutagenesis and production of mutant recombinant viruses demonstrated that both the Cys-181 and Cys-188 mutations cause resistance to E-EPSeU and E-EPU. Of the two mutations, the Cys-188 substitution produced greater E-EPSeU and E-EPU resistance. The predominance of the Cys-188 mutation in E-EPSeU-resistant variants has not been noted for other classes of HIV-1 specific RT inhibitors. HIV-1 resistance is likely to limit the therapeutic efficacy of acyclic 6-substituted pyrimidines if they are used as monotherapy.


Assuntos
Antivirais/farmacologia , HIV-1/efeitos dos fármacos , Compostos Organosselênicos/farmacologia , Uracila/análogos & derivados , Resistência a Medicamentos/genética , HIV-1/genética , Humanos , Mutação , Relação Estrutura-Atividade , Uracila/farmacologia , Zidovudina/farmacologia
10.
J Med Chem ; 37(16): 2583-8, 1994 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-8057301

RESUMO

A novel 5-o-carboranyl-containing nucleoside, 5-o-carboranyl-1-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)uracil (6, CFAU), was synthesized as a potential intracellular neutron capture agent. This compound was prepared in five steps starting from 5-iodo-1-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)uracil (1). The desired carboranyl derivative was obtained by addition of decaborane [as the bis(propionitrile) adduct] to the protected acetylenic nucleoside precursor followed by debenzoylation. The synthesis of CFAU was also performed by glycosylation of the suitably protected 5-o-carboranyluracil with the appropriate 2-fluoroarabinosyl derivative. This compound was evaluated for its cytotoxicity in human lymphocytes, monkey cells, and rat and human gliomas cells, as well as for antiviral activity against human immunodeficiency virus and herpes simplex virus type 1. Its biological activity was compared to 5-o-carboranyl-1-(2-deoxyribofuranosyl)uracil in these cell culture systems, human bone marrow cells, and mice. The results obtained to date suggest that CFAU has suitable characteristics as a sensitizer for boron neutron capture therapy.


Assuntos
Antivirais/síntese química , Arabinofuranosiluracila/análogos & derivados , Compostos de Boro/síntese química , Terapia por Captura de Nêutron de Boro , Radiossensibilizantes/síntese química , Animais , Antivirais/farmacologia , Arabinofuranosiluracila/síntese química , Arabinofuranosiluracila/farmacologia , Arabinofuranosiluracila/toxicidade , Medula Óssea/efeitos dos fármacos , Compostos de Boro/farmacologia , Compostos de Boro/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Feminino , Glioma/patologia , Glicosilação , HIV-1/efeitos dos fármacos , Herpesvirus Humano 1/efeitos dos fármacos , Humanos , Linfócitos/efeitos dos fármacos , Camundongos , Ratos , Células Tumorais Cultivadas
11.
Int J Radiat Oncol Biol Phys ; 28(5): 1113-20, 1994 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-8175396

RESUMO

PURPOSE: The intracellular uptake and metabolism of 5-carboranyl-2'-deoxyuridine was investigated in primary human lymphocytes and in a T lymphoblastoid cell line using unlabeled and tritium labeled compound. The cytotoxicity and antiviral activity of the compound and stability to enzyme degradation was determined. METHODS AND MATERIALS: A novel method for radiolabeling the 5-carboranyl moiety of pyrimidine nucleosides was developed. Cells were exposed to unlabeled and tritium labeled 5-carboranyl-2'-deoxyuridine and the intracellular uptake and egress of the compound determined by high pressure liquid chromatography. The viability and growth of normal and malignant cells, including human and rat gliomas, in the presence of the compound was determined. RESULTS: Substantial levels of 5-carboranyl-2'-deoxyuridine-5'-monophosphate are formed intracellularly and this major metabolite can be detected in cells 48 h after removal of the parent compound from the medium. No significant phosphorylation to the 5'-diphosphate or triphosphate of 5-carboranyl-2'-deoxyuridine was detected. Furthermore, radiolabeled 5-carboranyl-2'-deoxyuridine was not incorporated into deoxyribonucleic acid. 5-carboranyl-2'-deoxyuridine was essentially nontoxic to human lymphocytes as well as human or rat glioma cells, and had no marked effect in human lymphocytes acutely infected with human immunodeficiency virus type 1. CONCLUSION: The results demonstrate for the first time that 5-carboranyl-2'-deoxyuridine is phosphorylated intracellularly and suggest that it should be considered for further studies as a potential sensitizer for boron neutron capture therapy.


Assuntos
Compostos de Boro/metabolismo , Terapia por Captura de Nêutron de Boro , Desoxiuridina/análogos & derivados , Radiossensibilizantes/metabolismo , Compostos de Boro/síntese química , Compostos de Boro/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Desoxiuridina/síntese química , Desoxiuridina/metabolismo , Desoxiuridina/farmacologia , Humanos , Fosforilação
12.
J Med Chem ; 36(26): 4250-4, 1993 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-8277507

RESUMO

Lithiation of 5-bromo-2,4-bis(benzyloxy)pyrimidine (3) with n-BuLi at -80 degrees C followed by the addition of diphenyl diselenide or diphenyl disulfide as an electrophile furnished the corresponding 5-(phenylhetera)-2,4-bis(benzyloxy)pyrimidine, which on exposure to trimethylsilyl iodide in CH2-Cl2 at room temperature yielded the 5-(phenylhetera)uracils in 70-75% yield. Similarly, the 6-(phenylhetera)uracils were prepared from 6-bromo-2,4-bis(benzyloxy)pyrimidine (10). 1-[(2-Hydroxyethoxy)methyl]-5-(phenylselenenyl)uracil (PSAU, 18) and 1-(ethoxymethyl)-5-(phenylselenenyl)uracil (17) were synthesized by the electrophilic addition of benzeneselenenyl chloride to the acyclic uracils under basic conditions. These compounds were evaluated for their ability to inhibit dihydrouracil dehydrogenase (DHUDase, E.C. 1.3.1.2), orotate phosphoribosyltransferase (OPRTase, E.C. 2.4.2.10), uridine phosphorylase (UrdPase, E.C. 2.4.2.3), and thymidine phosphorylase (dThdPase, E.C. 2.4.2.4). 5-(Phenylselenenyl)uracil (PSU, 6) and 5-(phenylthio)uracil (PTU, 7) inhibited DHUDase with apparent K(i) values of 4.8 and 5.4 microM, respectively. The corresponding 6-analogues, compounds 13 and 14, demonstrated inhibitory activity against OPRTase. PTU as well as PSU and its riboside, 2'-deoxyriboside, and acyclonucleosides were inhibitors of UrdPase, with PSAU (18) being the most potent with an apparent K(i) value of 3.8 microM. None of the compounds evaluated had any effect on dThdPase. Interestingly, most of the compounds showed modest selective anti-human-immunodeficiency-virus activity in acutely infected primary human lymphocytes.


Assuntos
Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Oxirredutases/antagonistas & inibidores , Pirimidinas/química , Uridina Fosforilase/antagonistas & inibidores , Animais , Sobrevivência Celular/efeitos dos fármacos , Di-Hidrouracila Desidrogenase (NAD+) , Dissulfetos/química , Feminino , HIV-1/efeitos dos fármacos , Humanos , Fígado/enzimologia , Linfócitos/microbiologia , Camundongos , Compostos Organosselênicos/síntese química , Compostos Organosselênicos/química , Compostos Organosselênicos/farmacologia , Pirimidinas/síntese química , Pirimidinas/farmacologia , Selênio/química , Uracila/análogos & derivados , Uracila/síntese química , Uracila/química , Uracila/farmacologia
13.
Biochem Pharmacol ; 44(5): 973-80, 1992 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-1326966

RESUMO

3'-Azido-2',3'-dideoxyuridine (AzddU, CS-87) is a potent inhibitor of human immunodeficiency virus replication in vitro with low bone marrow toxicity. Although AzddU is currently being evaluated in clinical trials, its catabolic disposition is unknown. Pharmacokinetic studies in rhesus monkeys have demonstrated that a 5'-O-glucuronide is excreted in urine. The present study examined the catabolic disposition of AzddU is isolated rat hepatocytes, a model for the study at the cellular level of biosynthetic, catabolic and transport phenomena in the liver. Following exposure of cells to 10 microM [3H]AzddU, low intracellular levels of two catabolites, identified as 3'-azido-2',3'-dideoxy-5'-beta-D-glucopyranosyluridine (GAzddU) and 3'-amino-2',3'-dideoxyuridine (AMddU), were detected. Studies using rat microsomes demonstrated that GAzddU formation was only detected in the presence of uridine 5'-diphosphoglucuronic acid, and that the rate of AMddU formation increased significantly in the presence of NADPH. Under similar conditions, reduction of the 3'-azido function was also demonstrated herein with 3'-azido-2',3'-dideoxycytidine (AzddC), 3'-azido-2',3'-dideoxy-5-methylcytidine (AzddMeC) and 3'-azido-2',3'-dideoxyguanine (AzddG), suggesting that enzymatic reduction to a 3'-amino derivative is a general catabolic pathway of 3'-azido-2',3'-dideoxynucleosides at the hepatic site.


Assuntos
Antivirais/metabolismo , Desoxiuridina/análogos & derivados , Didesoxinucleosídeos/metabolismo , Microssomos Hepáticos/metabolismo , Zidovudina/análogos & derivados , Animais , Azidas/síntese química , Azidas/metabolismo , Cromatografia Líquida de Alta Pressão , Desoxiuridina/metabolismo , Desoxiuridina/farmacologia , Didesoxinucleosídeos/farmacologia , Glucuronatos/metabolismo , Células-Tronco Hematopoéticas/efeitos dos fármacos , NADP/farmacologia , Oxirredução , Ratos , Ratos Endogâmicos , Ribonucleosídeos , Uridina Difosfato Ácido Glucurônico/farmacologia , Zalcitabina/análogos & derivados , Zalcitabina/síntese química , Zalcitabina/metabolismo , Zidovudina/metabolismo
14.
J Med Chem ; 34(11): 3305-9, 1991 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1720175

RESUMO

Several 6-phenylselenenyl-substituted acyclouridine derivatives were prepared for evaluation as antiviral agents. Lithiation of the tert-butyldimethylsilyl-protected acyclonucleosides 4a-f with lithium diisopropylamide at -78 degrees C, followed by reaction with diphenyl diselenide as an electrophile, and subsequent removal of the protecting group with tetra n-butylammonium fluoride gave 1-[(2-hydroxyethoxy)methyl]-6-(phenylselenenyl)uracils 6a-f in 50-70% overall yield. The potency and spectrum of activity of compounds 6a-f against HIV-1 in vitro was similar to HEPT (1), a related 6-phenylthio acyclonucleoside. However, whereas HEPT inhibited HIV-1 reverse transcriptase, the selenium-containing derivatives were ineffective, suggesting a different mechanism of action. Of significance was the finding that the 6-phenylselenenyl acyclonucleosides inhibited also HIV-2 in primary human lymphocytes.


Assuntos
Antivirais/síntese química , HIV-1/efeitos dos fármacos , Nucleosídeos de Pirimidina/síntese química , Animais , Antivirais/farmacologia , HIV-2/efeitos dos fármacos , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/microbiologia , Nucleosídeos de Pirimidina/farmacologia , DNA Polimerase Dirigida por RNA/efeitos dos fármacos , Relação Estrutura-Atividade , Células Vero/efeitos dos fármacos
15.
Mol Pharmacol ; 39(2): 258-66, 1991 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-1996084

RESUMO

Metabolic studies in humans have demonstrated that 3'-azido-3'-deoxythymidine (AZT) is primarily eliminated as its 5'-O-glucuronide (GAZT). However, no detailed cellular metabolic studies have been reported on the complete catabolic fate of AZT at the hepatic site. Because the liver is probably the major site of AZT catabolism, the metabolism and transmembrane distribution of AZT were evaluated in freshly isolated rat hepatocytes, a model for the study at the cellular level of biosynthetic, catabolic, and transport phenomena in the liver. Following exposure of cells to 10 microM [3H]AZT, the predominant intracellular catabolite was GAZT, which reached a concentration of approximately 22 microM by 60 min. Additionally, under nonreducing conditions substantial levels of two previously unidentified AZT catabolites that were formed at the hepatic site and were distinct from any known anabolites or catabolites were also detected. These catabolites were identified as 3'-amino-3'-deoxythymidine (AMT) by fast atom bombardment mass spectrometry and 3'-amino-3'-deoxythymidine glucuronide (GAMT) through specific enzymatic hydrolysis. However, AMT was not a substrate for uridine 5'-diphosphoglucuronyltransferase and GAMT was found to be a reductive product of GAZT. Studies using rat and human liver microsomes demonstrated that the rate of formation of AMT and GAMT increased in the presence of NADPH, suggesting the involvement of a NADPH-dependent enzyme system. Studies using human hematopoietic progenitor cells demonstrated that AMT was 5- to 7-fold more toxic to human colony-forming units granulocyte-macrophage and burst-forming units erythroid than was AZT. This study provides the first detailed catabolic profile of AZT at the hepatic site and emphasizes the critical role that the liver plays in drug clearance. Formation of AMT, a highly toxic catabolite of AZT, raises a question regarding the role of AMT in the cytotoxic effects of AZT observed in patients.


Assuntos
Medula Óssea/efeitos dos fármacos , Didesoxinucleosídeos/metabolismo , Fígado/metabolismo , Microssomos Hepáticos/metabolismo , Zidovudina/metabolismo , Animais , Células da Medula Óssea , Didesoxinucleosídeos/toxicidade , Espaço Extracelular/metabolismo , Cromatografia Gasosa-Espectrometria de Massas/métodos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Líquido Intracelular/metabolismo , Fígado/citologia , Fígado/ultraestrutura , Ratos , Ratos Endogâmicos , Fatores de Tempo , Trítio , Zidovudina/análogos & derivados , Zidovudina/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...