Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Drug Metab Dispos ; 44(8): 1229-45, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27298339

RESUMO

The drug-metabolizing enzymes that contribute to the metabolism or bioactivation of a drug play a crucial role in defining the absorption, distribution, metabolism, and excretion properties of that drug. Although the overall effect of the cytochrome P450 (P450) family of drug-metabolizing enzymes in this capacity cannot be understated, advancements in the field of non-P450-mediated metabolism have garnered increasing attention in recent years. This is perhaps a direct result of our ability to systematically avoid P450 liabilities by introducing chemical moieties that are not susceptible to P450 metabolism but, as a result, may introduce key pharmacophores for other drug-metabolizing enzymes. Furthermore, the effects of both P450 and non-P450 metabolism at a drug's site of therapeutic action have also been subject to increased scrutiny. To this end, this Special Section on Emerging Novel Enzyme Pathways in Drug Metabolism will highlight a number of advancements that have recently been reported. The included articles support the important role of non-P450 enzymes in the clearance pathways of U.S. Food and Drug Administration-approved drugs over the past 10 years. Specific examples will detail recent reports of aldehyde oxidase, flavin-containing monooxygenase, and other non-P450 pathways that contribute to the metabolic, pharmacokinetic, or pharmacodynamic properties of xenobiotic compounds. Collectively, this series of articles provides additional support for the role of non-P450-mediated metabolic pathways that contribute to the absorption, distribution, metabolism, and excretion properties of current xenobiotics.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Xenobióticos/farmacocinética , Ativação Metabólica , Animais , Glucuronosiltransferase/metabolismo , Humanos , Inativação Metabólica , Oxirredução , Oxirredutases/metabolismo , Especificidade por Substrato , Sulfotransferases/metabolismo , Xenobióticos/efeitos adversos
2.
Drug Metab Dispos ; 44(8): 1262-9, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27079250

RESUMO

N1-Substituted-6-arylthiouracils, represented by compound 1 [6-(2,4-dimethoxyphenyl)-1-(2-hydroxyethyl)-2-thioxo-2,3-dihydropyrimidin-4(1H)-one], are a novel class of selective irreversible inhibitors of human myeloperoxidase. The present account is a summary of our in vitro studies on the facile oxidative desulfurization in compound 1 to a cyclic ether metabolite M1 [5-(2,4-dimethoxyphenyl)-2,3-dihydro-7H-oxazolo[3,2-a]pyrimidin-7-one] in NADPH-supplemented rats (t1/2 [half-life = mean ± S.D.] = 8.6 ± 0.4 minutes) and dog liver microsomes (t1/2 = 11.2 ± 0.4 minutes), but not in human liver microsomes (t1/2 > 120 minutes). The in vitro metabolic instability also manifested in moderate-to-high plasma clearances of the parent compound in rats and dogs with significant concentrations of M1 detected in circulation. Mild heat deactivation of liver microsomes or coincubation with the flavin-containing monooxygenase (FMO) inhibitor imipramine significantly diminished M1 formation. In contrast, oxidative metabolism of compound 1 to M1 was not inhibited by the pan cytochrome P450 inactivator 1-aminobenzotriazole. Incubations with recombinant FMO isoforms (FMO1, FMO3, and FMO5) revealed that FMO1 principally catalyzed the conversion of compound 1 to M1. FMO1 is not expressed in adult human liver, which rationalizes the species difference in oxidative desulfurization. Oxidation by FMO1 followed Michaelis-Menten kinetics with Michaelis-Menten constant, maximum rate of oxidative desulfurization, and intrinsic clearance values of 209 µM, 20.4 nmol/min/mg protein, and 82.7 µl/min/mg protein, respectively. Addition of excess glutathione essentially eliminated the conversion of compound 1 to M1 in NADPH-supplemented rat and dog liver microsomes, which suggests that the initial FMO1-mediated S-oxygenation of compound 1 yields a sulfenic acid intermediate capable of redox cycling to the parent compound in a glutathione-dependent fashion or undergoing further oxidation to a more electrophilic sulfinic acid species that is trapped intramolecularly by the pendant alcohol motif in compound 1.


Assuntos
Inibidores Enzimáticos/farmacocinética , Fígado/enzimologia , Oxigenases/metabolismo , Peroxidase/antagonistas & inibidores , Tiouracila/farmacocinética , Administração Intravenosa , Animais , Biotransformação , Cães , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/sangue , Meia-Vida , Humanos , Masculino , Taxa de Depuração Metabólica , Microssomos Hepáticos/enzimologia , Modelos Biológicos , Oxirredução , Oxigenases/antagonistas & inibidores , Peroxidase/metabolismo , Ratos Wistar , Especificidade da Espécie , Tiouracila/administração & dosagem , Tiouracila/análogos & derivados , Tiouracila/sangue
3.
Drug Metab Dispos ; 42(11): 1926-39, 2014 11.
Artigo em Inglês | MEDLINE | ID: mdl-25142735

RESUMO

The present article summarizes Metabolites in Safety Testing (MIST) studies on a glucokinase activator, N,N-dimethyl-5-((2-methyl-6-((5-methylpyrazin-2-yl)carbamoyl)benzofuran-4-yl)oxy)pyrimidine-2-carboxamide (PF-04937319), which is under development for the treatment of type 2 diametes mellitus. Metabolic profiling in rat, dog, and human hepatocytes revealed that PF-04937319 is metabolized via oxidative (major) and hydrolytic pathways (minor). N-Demethylation to metabolite M1 [N-methyl-5-((2-methyl-6-((5-methylpyrazin-2-yl)carbamoyl)benzofuran-4-yl)oxy)pyrimidine-2-carboxamide] was the major metabolic fate of PF-04937319 in human (but not rat or dog) hepatocytes, and was catalyzed by CYP3A and CYP2C isoforms. Qualitative examination of circulating metabolites in humans at the 100- and 300-mg doses from a 14-day multiple dose study revealed unchanged parent drug and M1 as principal components. Because M1 accounted for 65% of the drug-related material at steady state, an authentic standard was synthesized and used for comparison of steady-state exposures in humans and the 3-month safety studies in rats and dogs at the no-observed-adverse-effect level. Although circulating levels of M1 were very low in beagle dogs and female rats, adequate coverage was obtained in terms of total maximal plasma concentration (∼7.7× and 1.8×) and area under the plasma concentration-time curve (AUC; 3.6× and 0.8× AUC) relative to the 100- and 300-mg doses, respectively, in male rats. Examination of primary pharmacology revealed M1 was less potent as a glucokinase activator than the parent drug (compound PF-04937319: EC50 = 0.17 µM; M1: EC50 = 4.69 µM). Furthermore, M1 did not inhibit major human P450 enzymes (IC50 > 30 µM), and was negative in the Salmonella Ames assay, with minimal off-target pharmacology, based on CEREP broad ligand profiling. Insights gained from this analysis should lead to a more efficient and focused development plan for fulfilling MIST requirements with PF-04937319.


Assuntos
Benzofuranos/farmacocinética , Ativadores de Enzimas/farmacocinética , Glucoquinase/metabolismo , Pirimidinas/farmacocinética , Animais , Área Sob a Curva , Benzofuranos/sangue , Cães , Ativadores de Enzimas/sangue , Feminino , Humanos , Pirimidinas/sangue , Ratos
4.
Drug Metab Dispos ; 42(5): 918-31, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24608633

RESUMO

The disposition of a single oral dose of 5 mg (100 µCi) of [(14)C]axitinib was investigated in fasted healthy human subjects (N = 8). Axitinib was rapidly absorbed, with a median plasma Tmax of 2.2 hours and a geometric mean Cmax and half-life of 29.2 ng/ml and 10.6 hours, respectively. The plasma total radioactivity-time profile was similar to that of axitinib but the AUC was greater, suggesting the presence of metabolites. The major metabolites in human plasma (0-12 hours), identified as axitinib N-glucuronide (M7) and axitinib sulfoxide (M12), were pharmacologically inactive, and with axitinib comprised 50.4%, 16.2%, and 22.5% of the radioactivity, respectively. In excreta, the majority of radioactivity was recovered in most subjects by 48 hours postdose. The median radioactivity excreted in urine, feces, and total recovery was 22.7%, 37.0%, and 59.7%, respectively. The recovery from feces was variable across subjects (range, 2.5%-60.2%). The metabolites identified in urine were M5 (carboxylic acid), M12 (sulfoxide), M7 (N-glucuronide), M9 (sulfoxide/N-oxide), and M8a (methylhydroxy glucuronide), accounting for 5.7%, 3.5%, 2.6%, 1.7%, and 1.3% of the dose, respectively. The drug-related products identified in feces were unchanged axitinib, M14/15 (mono-oxidation/sulfone), M12a (epoxide), and an unidentified metabolite, comprising 12%, 5.7%, 5.1%, and 5.0% of the dose, respectively. The proposed mechanism to form M5 involved a carbon-carbon bond cleavage via M12a, followed by rearrangement to a ketone intermediate and subsequent Baeyer-Villiger rearrangement, possibly through a peroxide intermediate. In summary, the study characterized axitinib metabolites in circulation and primary elimination pathways of the drug, which were mainly oxidative in nature.


Assuntos
Imidazóis/farmacocinética , Indazóis/farmacocinética , Inibidores de Proteínas Quinases/farmacocinética , Proteínas Tirosina Quinases/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Adulto , Axitinibe , Radioisótopos de Carbono , Cromatografia Líquida de Alta Pressão , Fezes/química , Humanos , Imidazóis/sangue , Imidazóis/metabolismo , Imidazóis/urina , Indazóis/sangue , Indazóis/metabolismo , Indazóis/urina , Espectroscopia de Ressonância Magnética , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Estrutura Molecular , Inibidores de Proteínas Quinases/sangue , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/urina
5.
Pharm Res ; 30(4): 932-50, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23417511

RESUMO

PURPOSE: To discover drugs lowering PrP(Sc) in prion-infected cultured neuronal cells that achieve high concentrations in brain to test in mouse models of prion disease and then treat people with these fatal diseases. METHODS: We tested 2-AMT analogs for EC50 and PK after a 40 mg/kg single dose and 40-210 mg/kg/day doses for 3 days. We calculated plasma and brain AUC, ratio of AUC/EC50 after dosing. We reasoned that compounds with high AUC/EC50 ratios should be good candidates going forward. RESULTS: We evaluated 27 2-AMTs in single-dose and 10 in 3-day PK studies, of which IND24 and IND81 were selected for testing in mouse models of prion disease. They had high concentrations in brain after oral dosing. Absolute bioavailability ranged from 27-40%. AUC/EC50 ratios after 3 days were >100 (total) and 48-113 (unbound). Stability in liver microsomes ranged from 30->60 min. Ring hydroxylated metabolites were observed in microsomes. Neither was a substrate for the MDR1 transporter. CONCLUSIONS: IND24 and IND81 are active in vitro and show high AUC/EC50 ratios (total and unbound) in plasma and brain. These will be evaluated in mouse models of prion disease.


Assuntos
Proteínas PrPSc/antagonistas & inibidores , Doenças Priônicas/tratamento farmacológico , Tiazóis/metabolismo , Tiazóis/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Área Sob a Curva , Disponibilidade Biológica , Encéfalo/metabolismo , Linhagem Celular , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Camundongos , Microssomos Hepáticos/metabolismo , Proteínas PrPSc/metabolismo , Isoformas de Proteínas/metabolismo , Solubilidade , Tiazóis/química , Tiazóis/farmacologia
6.
Med Res Rev ; 33(5): 985-1080, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23090860

RESUMO

The decline in approval of new drugs during the past decade has led to a close analysis of the drug discovery process. One of the main reasons for attrition is preclinical toxicity, frequently attributed to the generation of protein-reactive drug metabolites. In this review, we present a critique of such reactive metabolites and evaluate the evidence linking them to observed toxic effects. Methodology for the characterization of reactive metabolites has advanced greatly in recent years, and is summarized first. Next, we consider the inhibition of key metabolic enzymes by electrophilic metabolites, as well as unfavorable drug-drug interactions that may ensue. One important class of protein-reactive metabolites, not linked conclusively to a toxic event, is acyl glucuronides. Their properties are discussed in light of the safety characteristics of carboxylic acid containing drugs. Many adverse drug reactions (ADRs) are known collectively as idiosyncratic events, that is, not predictable from knowledge of the pharmacology and pharmacokinetics of the parent compound. Observed ADRs may take various forms. Specific organ injury, particularly of the liver, is the most direct: we examine this in some detail. Moving to the cellular level, we also consider the upregulation of induced cellular processes. The related, but distinct, issue of hypersensitivity or allergic reactions to drugs and their metabolites, possibly via the immune system, is considered next. Finally, we discuss the impact of such data on the drug discovery process, both through early detection of reactive metabolites and informed synthetic design, which eliminates unfavorable functionality from drug candidates.


Assuntos
Desenho de Fármacos , Preparações Farmacêuticas/metabolismo , Animais , Pesquisa Biomédica , Sistema Enzimático do Citocromo P-450 , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Glucuronídeos/metabolismo , Humanos
7.
Expert Opin Drug Discov ; 7(7): 561-81, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22607458

RESUMO

INTRODUCTION: The design of target-specific covalent inhibitors is conceptually attractive because of increased biochemical efficiency through covalency and increased duration of action that outlasts the pharmacokinetics of the agent. Although many covalent inhibitors have been approved or are in advanced clinical trials to treat indications such as cancer and hepatitis C, there is a general tendency to avoid them as drug candidates because of concerns regarding immune-mediated toxicity that can arise from indiscriminate reactivity with off-target proteins. AREAS COVERED: The review examines potential reason(s) for the excellent safety record of marketed covalent agents and advanced clinical candidates for emerging therapeutic targets. A significant emphasis is placed on proteomic techniques and chemical/biochemical reactivity assays that aim to provide a systematic rank ordering of pharmacologic selectivity relative to off-target protein reactivity of covalent inhibitors. EXPERT OPINION: While tactics to examine selective covalent modification of the pharmacologic target are broadly applicable in drug discovery, it is unclear whether the output from such studies can prospectively predict idiosyncratic immune-mediated drug toxicity. Opinions regarding an acceptable threshold of protein reactivity/body burden for a toxic electrophile and a non-toxic electrophilic covalent drug have not been defined. Increasing confidence in proteomic and chemical/biochemical reactivity screens will require a retrospective side-by-side profiling of marketed covalent drugs and electrophiles known to cause deleterious toxic effects via non-selective covalent binding.


Assuntos
Descoberta de Drogas , Inibidores Enzimáticos/efeitos adversos , Inibidores Enzimáticos/química , Terapia de Alvo Molecular , Animais , Inibidores Enzimáticos/farmacocinética , Glutationa/metabolismo , Humanos , Camundongos , Ligação Proteica , Relação Estrutura-Atividade
8.
Toxicol In Vitro ; 25(5): 1061-6, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21513789

RESUMO

Thiabendazole (TBZ), an antihelminthic and antifungal agent, is associated with a host of adverse effects including nephrotoxicity, hepatotoxicity, and teratogenicity. Bioactivation of the primary metabolite of TBZ, 5-hydroxythiabendazole, has been proposed to yield a reactive intermediate. Here we show that this reactive intermediate can be catalyzed by myeloperoxidase (MPO), a neutrophil-bourne peroxidase. Using a cell viability endpoint, we examined the toxicity of TBZ, 5OH-TBZ, and MPO-generated metabolites in cell-based models including primary rat proximal tubule epithelial cells, NRK-52E rat proximal tubule cells, and H9C2 rat myocardial cells. Timecourse experiments with MPO showed complete turnover of 5OH-TBZ within 15 min and a dramatic leftward shift in dose-response curves after 12h. After a 24h exposure in vitro, the LC(50) of this reactive intermediate was 23.3 ± 0.2 µM reduced from greater than 200 µM from 5OH-TBZ alone, an approximately 10-fold decrease. LC(50) values were equal in all cell types used. Comparison of lactate dehydrogenase leakage and caspase 3/7 activity revealed that cell death caused by the reactive intermediate is primarily associated with necrosis rather than apoptosis. This toxicity can be completely rescued via incubation with rutin, an inhibitor of MPO. These results suggest that MPO-mediated biotransformation of 5OH-TBZ yields a reactive intermediate which may play a role in TBZ-induced toxicity.


Assuntos
Antifúngicos/toxicidade , Neutrófilos/patologia , Peroxidase/metabolismo , Tiabendazol/análogos & derivados , Animais , Biotransformação , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/patologia , L-Lactato Desidrogenase/metabolismo , Masculino , Necrose , Neutrófilos/metabolismo , Ratos , Ratos Wistar , Rutina/farmacologia , Tiabendazol/toxicidade
9.
Chem Res Toxicol ; 20(11): 1649-57, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17907785

RESUMO

The primary pathway of clearance of the methylenedioxyphenyl-containing compound and selective serotonin reuptake inhibitor paroxetine in humans involves P450 2D6-mediated demethylenation to a catechol intermediate. The process of demethylenation also results in the mechanism-based inactivation of the P450 isozyme. While the link between P450 2D6 inactivation and pharmacokinetic interactions of paroxetine with P450 2D6 substrates has been firmly established, there is a disconnect in terms of paroxetine's excellent safety record despite the potential for bioactivation. In the present study, we have systematically assessed the NADPH-dependent covalent binding of [(3)H]paroxetine to human liver microsomes and S-9 preparations in the absence and presence of cofactors of the various phase II drug-metabolizing enzymes involved in the downstream metabolism/detoxification of the putative paroxetine-catechol intermediate. Incubation of [(3)H]paroxetine with human liver microsomes and S-9 preparations resulted in irreversible binding of radioactive material to macromolecules by a process that was NADPH-dependent. The addition of reduced glutathione (GSH) to the microsomal and S-9 incubations resulted in a dramatic reduction of covalent binding. Following incubations with NADPH- and GSH-supplemented human liver microsomes and S-9, three sulfydryl conjugates with MH(+) ions at 623 Da (GS1), 779 Da (GS2), and 928 Da (GS3), respectively, were detected by LC-MS/MS. The collision-induced dissociation spectra allowed an insight into the structure of the GSH conjugates, based on which, bioactivation pathways were proposed. The formation of GS 1 was consistent with Michael addition of GSH to the quinone derived from two-electron oxidation of paroxetine-catechol. GS 3 was formed by the addition of a second molecule of GSH to the quinone species obtained via the two-electron oxidation of GS 1. The mechanism of formation of GS 2 can be rationalized via (i) further two-electron oxidation of the catechol motif in GS 3 to the ortho-quinone, (ii) loss of a glutamic acid residue from one of the adducted GSH molecules, and (iii) condensation of a cysteine-NH 2 with an adjacent carbonyl of the ortho-quinone to yield an ortho-benzoquinoneimine structure. Inclusion of the catechol-O-methyltransferase cofactor S-adenosylmethionine (SAM) in S-9 incubations also dramatically reduced the covalent binding of [(3)H]paroxetine, a finding that was consistent with O-methylation of the paroxetine-catechol metabolite to the corresponding guaiacol regioisomers in S-9 incubations. While the NADPH-dependent covalent binding was attenuated by GSH and SAM, these reagents did not alter paroxetine's ability to inactivate P450 2D6, suggesting that the reactive intermediate responsible for P450 inactivation did not leave the active site to react with other proteins. The results of our studies indicate that in addition to the low once-a-day dosing regimen (20 mg) of paroxetine, efficient scavenging of the catechol and quinone metabolites by SAM and GSH, respectively, serves as an explanation for the excellent safety record of paroxetine despite the fact that it undergoes bioactivation.


Assuntos
Microssomos Hepáticos/metabolismo , NADP/metabolismo , Paroxetina/metabolismo , Quinonas/metabolismo , Biotransformação , Inibidores do Citocromo P-450 CYP2D6 , Glutationa/metabolismo , Humanos , Quinonas/química , Trítio
10.
Rapid Commun Mass Spectrom ; 21(20): 3317-22, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17879390

RESUMO

Carbamazepine (CBZ)-induced idiosyncratic toxicities are commonly believed to be related to the formation of reactive metabolites. CBZ is metabolized primarily into carbamazepine-10,11-epoxide (CBZE), 2-hydroxycarbamazepine (2-OHCBZ) and 3-hydroxycarbamazepine (3-OHCBZ), in human liver microsomes (HLM). Over the past two decades, the 2,3-arene oxidation has been commonly assumed to be the major bioactivation pathway of CBZ. Recently, CBZE has been also confirmed to be chemically reactive. In order to identify other possible primary and sequential CBZ bioactivation pathways, individual HLM incubations of CBZ, CBZE, 2-OHCBZ and 3-OHCBZ were conducted in the presence of glutathione (GSH). In the CBZ incubation, a variety of GSH adducts were formed via individual or combined pathways of 10,11-epoxidation, arene oxidation and iminoquinone formation. In the CBZE incubation, the only detected GSH adducts were CBZE-SG1 and CBZE-SG2, which represented the two most abundant conjugates observed in the CBZ incubation. In the incubation of either 2-OHCBZ or 3-OHCBZ, a number of sequential GSH adducts were observed. However, none of the 2-OHCBZ-derived GSH adducts were detected in the CBZ incubation. Meanwhile, several GSH adducts were only observed in the CBZ incubation. In conclusion, CBZ can be bioactivated in HLM via 10,11-epoxidation, 2,3-arene oxidation, and several other pathways. In addition, the sequential bioactivation of 3-OHCBZ appeared to play a more important role than that of either CBZE or 2-OHCBZ in the overall bioactivation of CBZ in HLM. The identification of several new bioactivation pathways of CBZ in HLM demonstrates that possible CBZ bioactivation can be more complex than previously thought.


Assuntos
Anticonvulsivantes/farmacocinética , Carbamazepina/farmacocinética , Cromatografia Líquida de Alta Pressão , Microssomos Hepáticos/metabolismo , Espectrometria de Massas por Ionização por Electrospray/métodos , Espectrometria de Massas em Tandem/métodos , Biotransformação , Glutationa/metabolismo , Humanos
11.
Drug Metab Dispos ; 35(6): 848-58, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17344339

RESUMO

2-(3-Chlorobenzyloxy)-6-(piperazin-1-yl)pyrazine (3) is a potent and selective 5-HT(2C) agonist that exhibits dose-dependent inhibition of food intake and reduction in body weight in rats, making it an attractive candidate for treatment of obesity. However, examination of the genotoxicity potential of 3 in the Salmonella Ames assay using tester strains TA98, TA100, TA1535, and TA1537 revealed a metabolism (rat S9/NADPH)- and dose-dependent increase of reverse mutations in strains TA100 and TA1537. The increase in reverse mutations was attenuated upon coincubation with methoxylamine and glutathione. The irreversible and concentration-dependent incorporation of radioactivity in calf thymus DNA after incubations with [14C]3 in the presence of rat S9/NADPH suggested that 3 was bioactivated to a reactive intermediate that covalently bound DNA. In vitro metabolism studies on 3 with rat S9/NADPH in the presence of methoxylamine and cyanide led to the detection of amine and cyano conjugates of 3. The mass spectrum of the amine conjugate was consistent with condensation of amine with an aldehyde metabolite derived from hydroxylation of the secondary piperazine nitrogen-alpha-carbon bond. The mass spectrum of the cyano conjugate suggested a bioactivation pathway involving N-hydroxylation of the secondary piperazine nitrogen followed by two-electron oxidation to generate an electrophilic nitrone, which reacted with cyanide. The 3-chlorobenzyl motif in 3 was also bioactivated via initial aromatic ring hydroxylation followed by elimination to a quinone-methide species that reacted with glutathione or with the secondary piperazine ring nitrogen in 3 and its monohydroxylated metabolite(s). The metabolism studies described herein provide a mechanistic basis for the mutagenicity of 3.


Assuntos
Piperazinas/toxicidade , Pirazinas/toxicidade , Salmonella typhimurium/efeitos dos fármacos , Agonistas do Receptor 5-HT2 de Serotonina , Biotransformação , Testes de Mutagenicidade , Obesidade , Piperazinas/síntese química , Piperazinas/metabolismo , Pirazinas/síntese química , Pirazinas/metabolismo , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo
12.
Drug Metab Dispos ; 34(8): 1310-6, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16679384

RESUMO

Nomifensine is an antidepressant agent that was removed from use because of a high incidence of hemolytic anemia. It contains an N-methyl-8-aminotetrahydroisoquinoline ring which has the potential to be oxidized to quaternary dihydroisoquinolinium and isoquinolinium ions, albeit such a transformation had not been previously observed. In this report, we demonstrate the conversion of nomifensine to a dihydroisoquinolinium ion metabolite by several human enzymes. Human liver microsomes supplemented with NADPH generated the dihydroisoquinolinium ion metabolite along with other hydroxylated metabolites, whereas when supplemented with t-butyl peroxide, only the dihydroisoquinolinium ion metabolite was observed. Monoamine oxidase A, but not monoamine oxidase B, catalyzed this reaction, as well as human hemoglobin supplemented with H2O2. Human myeloperoxidase catalyzed this reaction in the presence of H2O2, and activation of the reaction was observed when incubations were conducted in the presence of acetaminophen at concentrations relevant to those measured in humans. The reaction was also observed in human whole blood. The equilibrium between the dihydroisoquinolinium ion and carbinolamine was shown to have a pK of about 11.7. The dihydroisoquinolinium ion was shown to react with cyanide and borohydride, but not glutathione. These findings suggest that the electrophilic nomifensine dihydroisoquinolinium metabolite, which can be generated by several enzymes, could be behind toxic responses to nomifensine such as hemolytic anemia and hepatotoxicity.


Assuntos
Nomifensina/metabolismo , Compostos de Quinolínio/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Hemoglobinas/metabolismo , Humanos , Técnicas In Vitro , Microssomos Hepáticos/metabolismo , Monoaminoxidase/metabolismo , Peroxidase/metabolismo
13.
Curr Drug Metab ; 6(3): 161-225, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15975040

RESUMO

The occurrence of idiosyncratic adverse drug reactions during late clinical trials or after a drug has been released can lead to a severe restriction in its use and even in its withdrawal. Metabolic activation of relatively inert functional groups to reactive electrophilic intermediates is considered to be an obligatory event in the etiology of many drug-induced adverse reactions. Therefore, a thorough examination of the biochemical reactivity of functional groups/structural motifs in all new drug candidates is essential from a safety standpoint. A major theme attempted in this review is the comprehensive cataloging of all of the known bioactivation pathways of functional groups or structural motifs commonly utilized in drug design efforts. Potential strategies in the detection of reactive intermediates in biochemical systems are also discussed. The intention of this review is not to "black list" functional groups or to immediately discard compounds based on their potential to form reactive metabolites, but rather to serve as a resource describing the structural diversity of these functionalities as well as experimental approaches that could be taken to evaluate whether a "structural alert" in a new drug candidate undergoes bioactivation to reactive metabolites.


Assuntos
Preparações Farmacêuticas/química , Preparações Farmacêuticas/metabolismo , Animais , Biotransformação/fisiologia , Humanos , Microssomos Hepáticos/metabolismo , Compostos Orgânicos/química , Compostos Orgânicos/metabolismo , Relação Estrutura-Atividade
14.
Drug Metab Dispos ; 32(1): 49-57, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14709620

RESUMO

Ticlopidine is an agent that inhibits adenosine diphosphate-induced platelet aggregation. Metabolic studies with ticlopidine have indicated that the principal routes of metabolism are N-dealkylation, N-oxidation, and oxidation of the thiophene ring. However, ticlopidine shares some structural features that are similar to those of cyclic tertiary amines such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and tetrahydroisoquinolines, which are converted to neurotoxic pyridinium metabolites, via the iminium (dihydropyridinium) species. The current in vitro studies examined the potential of ticlopidine to undergo a similar conversion by cytochrome P450 (P450), peroxidases, and monoamine oxidase (MAO). The results from these studies have suggested that ticlopidine undergoes an overall 4-electron oxidation to the novel thienopyridinium metabolite (M6) via the intermediate 2-electron oxidation product, the thienodihydropyridinium metabolite (M5) by P450, horseradish peroxidase, and myeloperoxidase and, to a lesser extent, by MAO. The structures of these metabolites were characterized by liquid chromatography (LC)-tandem mass spectrometry and LC-NMR. Qualitative studies with baculovirus-expressed P450s revealed the involvement of P450 3A4 in this conversion. Interestingly, M5 was the primary metabolite in the peroxidase-mediated reactions and was quite stable to air oxidation or disproportionation. It was less electrophilic and did not form cyanide, glutathione, or N-acetylcysteine adducts. On the other hand, M6 was the major metabolite in P450-catalyzed oxidation of ticlopidine. The results from this study have revealed that in addition to metabolism of the thiophene ring of ticlopidine, the tetrahydropyridine moiety of the compound is susceptible to a 2-electron and a 4-electron oxidation like other cyclic tertiary amines.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Monoaminoxidase/metabolismo , Peroxidases/metabolismo , Inibidores da Agregação Plaquetária/farmacocinética , Compostos de Piridínio/metabolismo , Tiofenos/metabolismo , Ticlopidina/farmacocinética , Animais , Cromatografia Líquida de Alta Pressão , Sistema Enzimático do Citocromo P-450/biossíntese , Sistema Enzimático do Citocromo P-450/genética , DNA Complementar/biossíntese , Peroxidase do Rábano Silvestre/metabolismo , Humanos , Técnicas In Vitro , Insetos , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Microssomos Hepáticos , Monoaminoxidase/biossíntese , Monoaminoxidase/genética , Oxirredução , Peroxidases/biossíntese , Peroxidases/genética
15.
Drug Metab Dispos ; 31(11): 1369-77, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14570769

RESUMO

The nonsteroidal anti-inflammatory agent (+ or -)-suprofen [alpha-methyl-4-(2-thienylcarbonyl)benzeneacetic acid] was evaluated as a P450 2C9 inactivator. (+ or -)-Suprofen inactivated the diclofenac-4-hydroxylase activity of baculovirus-expressed P450 2C9 in a time- and concentration-dependent manner, which was consistent with mechanism-based inactivation. The loss of activity followed pseudo-first-order kinetics and was suprofen- and NADPH-dependent. The kinetic parameters for inactivation kinact and KI were 0.091 min-1 and 3.7 microM, respectively, and the partition ratio was 101. Although P450 2C9 substrate S-warfarin partially protected against inactivation, reactive oxygen scavengers such as superoxide dismutase and catalase did not prevent inactivation. Extensive dialysis did not regenerate enzyme activity, suggesting that inactivation proceeded via covalent modification. Inactivated P450 2C9 lost <10% of its ability to form a CO-reduced complex, suggesting that inactivation may have resulted from covalent modification of apoprotein. Addition of exogenous nucleophiles such as glutathione and semicarbazide partially protected against inactivation. Apart from the metabolism of suprofen to 5-hydroxysuprofen, the formation of a suprofen-glutathione conjugate was also discernible in microsomal mixtures containing glutathione. Time of flight mass spectrometry revealed a protonated monoisotopic mass of 566.1304 for this conjugate, consistent with an elemental composition of C24H28N3O9S2. The mass spectrum indicated that conjugation had occurred on the intact thiophene ring, presumably via a thioether linkage. Further evidence for the formation of an electrophilic intermediate in suprofen-P450 2C9 incubations was obtained via the characterization of a novel pyridazine adduct upon addition of semicarbazide to the microsomal mixtures. The pyridazine derivative had a protonated monoisotopic mass of 257.0895 that was consistent with an elemental composition of C14H13O3N2. The formation of the stable pyridazine adduct suggested the generation of an electrophilic gamma-thioketo-alpha, beta-unsaturated aldehyde, analogous to that observed during the cytochrome P450-mediated bioactivation of furan. This electrophilic alpha, beta-unsaturated aldehyde represents a possible reactive intermediate that bioalkylates P450 2C9.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Suprofeno/farmacologia , Anti-Inflamatórios não Esteroides/química , Hidrocarboneto de Aril Hidroxilases/metabolismo , Citocromo P-450 CYP2C9 , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Humanos , Microssomos/efeitos dos fármacos , Microssomos/enzimologia , Suprofeno/química
16.
Drug Metab Dispos ; 31(10): 1240-50, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12975333

RESUMO

The 3-unsubstituted isoxazole ring in the anti-inflammatory drug leflunomide undergoes a unique N-O bond cleavage to the active alpha-cyanoenol metabolite A771726, which resides in the same oxidation state as the parent. In vitro studies were conducted to characterize drug-metabolizing enzyme(s) responsible for ring opening and to gain insight into the mechanism of ring opening. Under physiological conditions, leflunomide was converted to A771726 in rat and human plasma (rat plasma,t1/2 = 36 min; human plasma, t1/2 = 12 min) and whole blood (rat blood, t1/2 = 59 min; human blood, t1/2 = 43 min). Human serum albumin also catalyzed A771726 formation, albeit at a much slower rate (t1/2 = 110 min). Rat and human liver microsomes also demonstrated NADPH-dependent A771726 formation (human liver microsomes, Vmax = 1797 pmol/min/mg and Km = 274 microM). Leflunomide metabolism in microsomes was sensitive to furafylline treatment, suggesting p4501A2 involvement. 3-Methylleflunomide, which contained a 3-methyl substituent on the isoxazole ring, was resistant to ring opening in base, plasma, blood, and liver microsomes. In microsomes, two monohydroxylated metabolites were formed, and metabolite identification studies established the 3- and the 5-methyl groups on the isoxazole ring as sites of hydroxylation. These results indicate that the C3-H in leflunomide is essential for ring opening. Although A771726 formation in human liver microsomes or recombinant p4501A2 required NADPH, its formation was greatly reduced by oxygen or carbon monoxide, suggesting that the isoxazole ring opening was catalyzed by the p450Fe(II) form of the enzyme. A mechanism for the p450-mediated ring scission is proposed in which the isoxazole ring nitrogen or oxygen coordinates to the reduced form of the heme followed by charge transfer from p450Fe(II) to the C=N bond or deprotonation of the C3-H, which results in a cleavage of the N-O bond.


Assuntos
Compostos de Anilina/metabolismo , Anti-Inflamatórios não Esteroides/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Hidroxibutiratos/metabolismo , Isoxazóis/metabolismo , Oximas/metabolismo , Compostos de Anilina/química , Animais , Anti-Inflamatórios não Esteroides/química , Crotonatos , Dessecação/métodos , Humanos , Hidroxibutiratos/química , Isoxazóis/química , Leflunomida , Masculino , Microssomos Hepáticos/metabolismo , Nitrilas , Oximas/química , Ratos , Ratos Sprague-Dawley , Toluidinas
17.
J Biol Chem ; 278(39): 37099-111, 2003 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-12842871

RESUMO

Inhibition of acetyl-CoA carboxylase (ACC), with its resultant inhibition of fatty acid synthesis and stimulation of fatty acid oxidation, has the potential to favorably affect the multitude of cardiovascular risk factors associated with the metabolic syndrome. To achieve maximal effectiveness, an ACC inhibitor should inhibit both the lipogenic tissue isozyme (ACC1) and the oxidative tissue isozyme (ACC2). Herein, we describe the biochemical and acute physiological properties of CP-610431, an isozyme-nonselective ACC inhibitor identified through high throughput inhibition screening, and CP-640186, an analog with improved metabolic stability. CP-610431 inhibited ACC1 and ACC2 with IC50s of approximately 50 nm. Inhibition was reversible, uncompetitive with respect to ATP, and non-competitive with respect to bicarbonate, acetyl-CoA, and citrate, indicating interaction with the enzymatic carboxyl transfer reaction. CP-610431 also inhibited fatty acid synthesis, triglyceride (TG) synthesis, TG secretion, and apolipoprotein B secretion in HepG2 cells (ACC1) with EC50s of 1.6, 1.8, 3.0, and 5.7 microm, without affecting either cholesterol synthesis or apolipoprotein CIII secretion. CP-640186, also inhibited both isozymes with IC50sof approximately 55 nm but was 2-3 times more potent than CP-610431 in inhibiting HepG2 cell fatty acid and TG synthesis. CP-640186 also stimulated fatty acid oxidation in C2C12 cells (ACC2) and in rat epitrochlearis muscle strips with EC50s of 57 nm and 1.3 microm. In rats, CP-640186 lowered hepatic, soleus muscle, quadriceps muscle, and cardiac muscle malonyl-CoA with ED50s of 55, 6, 15, and 8 mg/kg. Consequently, CP-640186 inhibited fatty acid synthesis in rats, CD1 mice, and ob/ob mice with ED50s of 13, 11, and 4 mg/kg, and stimulated rat whole body fatty acid oxidation with an ED50 of approximately 30 mg/kg. Taken together, These observations indicate that isozyme-nonselective ACC inhibition has the potential to favorably affect risk factors associated with the metabolic syndrome.


Assuntos
Acetil-CoA Carboxilase/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Ácidos Graxos/metabolismo , Isoenzimas/antagonistas & inibidores , Malonil Coenzima A/análise , Tecido Adiposo/metabolismo , Animais , Células Cultivadas , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C3H , Músculo Esquelético/metabolismo , Oxirredução , Ratos , Ratos Sprague-Dawley , Triglicerídeos/biossíntese
18.
Curr Drug Metab ; 3(4): 379-424, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12093357

RESUMO

The unexpected occurrence of idiosyncratic drug reactions during late clinical trials or after a drug has been released can lead to a severe restriction in its use or failure to release/withdrawal. This leads to considerable uncertainty in drug development and has led to attempts to try to predict a drug's potential to cause such reactions. The biotransformation of relatively inert drugs to highly reactive metabolites, commonly referred to as "bioactivation", is now recognized to be an obligatory step in several kinds of drug-induced adverse reactions. Reactive metabolites can be formed by most, if not all, of the enzymes that are involved in drug metabolism. A major theme explored in this review includes the diversity of oxidative bioactivation reactions on nitrogen-containing xenobiotics including drugs. A variety of Phase I enzymes including P450, MAO, and peroxidases bioactivate nitrogen-containing xenobiotics via direct oxidations on the nitrogen atom leading to reactive intermediates or by oxidation at an alternate site in the molecule; for the metabolite to be reactive via the latter sequence nitrogen participation in required. Examples of direct oxidations on nitrogen include the N-oxidation of aromatic amines (e.g. procainamide), single electron N-oxidation of imides (e.g. phenytoin), or alpha-carbon oxidations of arylalkyl- or alkylamines (e.g. mianserin), to reactive nitroso, nitrogen free radical and iminium species, respectively. Examples of indirect bioactivation are highlighted with aromatic amines (e.g. diclofenac) that undergo p-hydroxylation resulting in the formation of p-aminophenols, two-electron oxidation of which results in the formation of reactive quinoneimines. Potential strategies that could be utilized in the screening of novel bioactivation pathways are also discussed.


Assuntos
Compostos de Nitrogênio/metabolismo , Preparações Farmacêuticas/química , Preparações Farmacêuticas/metabolismo , Xenobióticos/química , Xenobióticos/metabolismo , Biotransformação , Estrutura Molecular , Oxirredução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...