Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Drug Metab Pharmacokinet ; 26(3): 266-79, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21383524

RESUMEN

The metabolism of 3,4-dihydro-7-[4-(1-naphthalenyl)-1-piperazinyl]butoxy]-1,8-naphthyridin-2(1H)-one (NPBN) was investigated in rats. Animals were administered 30 mg/kg NPBN that was labeled with both tritium and carbon-14. The mass recovery of drug-related material was 96-98%, with almost all material excreted in feces. Metabolism occurred by oxidation reactions followed by conjugation. The main route of metabolism of NPBN occurred via oxidation of the naphthylene ring, which led to naphthol and dihydrodiol metabolites as well as a relatively novel N-dearylated metabolite in which the naphthylene ring was removed. In vitro investigation in rat liver microsomes also showed a glutathione adduct on the naphthalene and a glutathione adduct of naphthoquinone, which, along with the dihydrodiol metabolite, is consistent with the initial generation of an epoxide. A mechanism is proposed whereby the N-dearylation arises via epoxidation, followed by formation of an exocyclic iminium ion intermediate that is hydrolyzed to yield the N-dearylated metabolite. An additional mechanism involves oxidation of the naphthol metabolite via a radical mechanism, since this metabolite was also shown to undergo N-dearylation.


Asunto(s)
Antipsicóticos/metabolismo , Antagonistas de Dopamina/metabolismo , Antagonistas de los Receptores de Dopamina D2 , Agonismo Parcial de Drogas , Animales , Antipsicóticos/sangre , Antipsicóticos/farmacocinética , Antipsicóticos/orina , Área Bajo la Curva , Cromatografía Líquida de Alta Presión , Remoción de Radical Alquila , Antagonistas de Dopamina/sangre , Antagonistas de Dopamina/farmacocinética , Antagonistas de Dopamina/orina , Compuestos Epoxi/metabolismo , Heces/química , Femenino , Glucurónidos/sangre , Glucurónidos/metabolismo , Glutatión/análogos & derivados , Glutatión/metabolismo , Hidroxilación , Espectroscopía de Resonancia Magnética , Masculino , Microsomas Hepáticos/metabolismo , Estructura Molecular , Naftiridinas/sangre , Naftiridinas/metabolismo , Naftiridinas/farmacocinética , Naftiridinas/orina , Oxidación-Reducción , Piperazinas/sangre , Piperazinas/metabolismo , Piperazinas/farmacocinética , Piperazinas/orina , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Ionización de Electrospray , Ésteres del Ácido Sulfúrico/metabolismo , Orina/química
2.
Chem Res Toxicol ; 23(6): 1115-26, 2010 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-20507089

RESUMEN

The synthesis and structure-activity relationship studies on 5-trifluoromethylpyrido[4,3-d]pyrimidin-4(3H)-ones as antagonists of the human calcium receptor (CaSR) have been recently disclosed [ Didiuk et al. ( 2009 ) Bioorg. Med. Chem. Lett. 19 , 4555 - 4559 ). On the basis of its pharmacology and disposition attributes, (R)-2-(2-hydroxyphenyl)-3-(1-phenylpropan-2-yl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one (1) was considered for rapid advancement to first-in-human (FIH) trials to mitigate uncertainty surrounding the pharmacokinetic/pharmacodynamic (PK/PD) predictions for a short-acting bone anabolic agent. During the course of metabolic profiling, however, glutathione (GSH) conjugates of 1 were detected in human liver microsomes in an NADPH-dependent fashion. Characterization of the GSH conjugate structures allowed insight(s) into the bioactivation pathway, which involved CYP3A4-mediated phenol ring oxidation to the catechol, followed by further oxidation to the electrophilic ortho-quinone species. While the reactive metabolite (RM) liability raised concerns around the likelihood of a potential toxicological outcome, a more immediate program goal was establishing confidence in human PK predictions in the FIH study. Furthermore, the availability of a clinical biomarker (serum parathyroid hormone) meant that PD could be assessed side by side with PK, an ideal scenario for a relatively unprecedented pharmacologic target. Consequently, progressing 1 into the clinic was given a high priority, provided the compound demonstrated an adequate safety profile to support FIH studies. Despite forming identical RMs in rat liver microsomes, no clinical or histopathological signs prototypical of target organ toxicity were observed with 1 in in vivo safety assessments in rats. Compound 1 was also devoid of metabolism-based mutagenicity in in vitro (e.g., Salmonella Ames) and in vivo assessments (micronuclei induction in bone marrow) in rats. Likewise, metabolism-based studies (e.g., evaluation of detoxicating routes of clearance and exhaustive PK/PD studies in animals to prospectively predict the likelihood of a low human efficacious dose) were also conducted, which mitigated the risks of idiosyncratic toxicity to a large degree. In parallel, medicinal chemistry efforts were initiated to identify additional compounds with a complementary range of human PK predictions, which would maximize the likelihood of achieving the desired PD effect in the clinic. The back-up strategy also incorporated an overarching goal of reducing/eliminating reactive metabolite formation observed with 1. Herein, the collective findings from our discovery efforts in the CaSR program, which include the incorporation of appropriate derisking steps when dealing with RM issues are summarized.


Asunto(s)
Anabolizantes/química , Anabolizantes/metabolismo , Osteoporosis/tratamiento farmacológico , Piridinas/química , Piridinas/metabolismo , Pirimidinonas/química , Pirimidinonas/metabolismo , Receptores Sensibles al Calcio/antagonistas & inhibidores , Anabolizantes/efectos adversos , Animales , Cristalografía por Rayos X , Humanos , Piridinas/efectos adversos , Pirimidinonas/efectos adversos , Ratas
3.
Chem Res Toxicol ; 22(2): 332-40, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19161298

RESUMEN

In vitro covalent binding studies in which xenobiotics are shown to undergo metabolism-dependent covalent binding to macromolecules have been commonly used to shed light on the biochemical mechanisms of xenobiotic-induced toxicity. In this paper, 18 drugs (nine hepatotoxins and nine nonhepatotoxins) were tested for their proclivity to demonstrate metabolism-dependent covalent binding to macromolecules in human liver S-9 fraction (9000 g supernatant) or human hepatocytes, as an extension to previous work that used human liver microsomes published in this journal [ Obach et al. ( 2008 ) Chem. Res. Toxicol. 21 , 1814 -1822 ]. In the S-9 fraction, seven out of the nine drugs in each category demonstrated some level of metabolism-dependent covalent binding. Inclusion of reduced glutathione, cofactors needed by conjugating enzymes, and other parameters (total daily dose and fraction of total intrinsic clearance comprised by covalent binding) improved the ability of the system to separate hepatotoxins from nonhepatotoxins to a limited extent. Covalent binding in human hepatocytes showed that six out of the nine hepatotoxins and four out of eight nonhepatotoxins demonstrated covalent binding. Taking into account estimates of total daily body burden of covalent binding from the hepatocyte data showed an improvement over other in vitro systems for distinguishing hepatotoxins from nonhepatotoxins; however, this metabolism system still displayed some false positives. Combined with the previous study using liver microsomes, these findings identify the limitations of in vitro covalent binding data for prospective prediction of hepatotoxicity for new drug candidates and highlight the need for a better understanding of the link between drug bioactivation, covalent adduct formation, and toxicity outcomes. Directly relating covalent binding to hepatotoxicity is likely an oversimplification of the process whereby adduct formation ultimately leads to toxicity. Understanding underlying complexities (e.g., which macromolecules are important covalent binding targets, interindividual differences in susceptibility, etc.) will be essential to any understanding of the problem of metabolism-dependent hepatotoxicity and predicting toxicity from in vitro experiments.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Hepatocitos/metabolismo , Preparaciones Farmacéuticas/metabolismo , Pruebas de Toxicidad/métodos , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Hepatocitos/efectos de los fármacos , Humanos , Microsomas Hepáticos/química , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Preparaciones Farmacéuticas/química , Toxinas Biológicas/metabolismo , Xenobióticos/metabolismo , Xenobióticos/toxicidad
4.
Curr Drug Metab ; 9(9): 847-53, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18991580

RESUMEN

Evaluation and optimization of drug metabolism and pharmacokinetic data plays an important role in drug discovery and development and several reliable in vitro ADME models are available. Recently higher throughput in vitro ADME screening facilities have been established in order to be able to evaluate an appreciable fraction of synthesized compounds. The ADME screening process can be dissected in five distinct steps: (1) plate management of compounds in need of in vitro ADME data, (2) optimization of the MS/MS method for the compounds, (3) in vitro ADME experiments and sample clean up, (4) collection and reduction of the raw LC-MS/MS data and (5) archival of the processed ADME data. All steps will be described in detail and the value of the data on drug discovery projects will be discussed as well. Finally, in vitro ADME screening can generate large quantities of data obtained under identical conditions to allow building of reliable in silico models.


Asunto(s)
Evaluación Preclínica de Medicamentos/normas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Preparaciones Farmacéuticas/metabolismo , Animales , Simulación por Computador , Evaluación Preclínica de Medicamentos/estadística & datos numéricos , Humanos , Farmacocinética , Control de Calidad
5.
Bioorg Med Chem Lett ; 18(23): 6071-7, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18951788

RESUMEN

The synthesis and SAR for a series of diaminopyrimidines as PYK2 inhibitors are described. Using a combination of library and traditional medicinal chemistry techniques, a FAK-selective chemical series was transformed into compounds possessing good PYK2 potency and 10- to 20-fold selectivity against FAK. Subsequent studies found that the majority of the compounds were positive in a reactive metabolite assay, an indicator for potential toxicological liabilities. Based on the proposed mechanism for bioactivation, as well as a combination of structure-based drug design and traditional medicinal chemistry techniques, a follow-up series of PYK2 inhibitors was identified that maintained PYK2 potency, FAK selectivity and HLM stability, yet were negative in the RM assay.


Asunto(s)
Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Pirimidinas/síntesis química , Pirimidinas/farmacología , Animales , Técnicas Químicas Combinatorias , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Diseño de Fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Humanos , Conformación Molecular , Estructura Molecular , Osteoporosis/tratamiento farmacológico , Pirimidinas/química , Ratas , Relación Estructura-Actividad
6.
Chem Res Toxicol ; 21(9): 1814-22, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18690722

RESUMEN

In vitro covalent binding assessments of drugs have been useful in providing retrospective insights into the association between drug metabolism and a resulting toxicological response. On the basis of these studies, it has been advocated that in vitro covalent binding to liver microsomal proteins in the presence and the absence of NADPH be used routinely to screen drug candidates. However, the utility of this approach in predicting toxicities of drug candidates accurately remains an unanswered question. Importantly, the years of research that have been invested in understanding metabolic bioactivation and covalent binding and its potential role in toxicity have focused only on those compounds that demonstrate toxicity. Investigations have not frequently queried whether in vitro covalent binding could be observed with drugs with good safety records. Eighteen drugs (nine hepatotoxins and nine nonhepatotoxins in humans) were assessed for in vitro covalent binding in NADPH-supplemented human liver microsomes. Of the two sets of nine drugs, seven in each set were shown to undergo some degree of covalent binding. Among hepatotoxic drugs, acetaminophen, carbamazepine, diclofenac, indomethacin, nefazodone, sudoxicam, and tienilic acid demonstrated covalent binding, while benoxaprofen and felbamate did not. Of the nonhepatotoxic drugs evaluated, buspirone, diphenhydramine, meloxicam, paroxetine, propranolol, raloxifene, and simvastatin demonstrated covalent binding, while ibuprofen and theophylline did not. A quantitative comparison of covalent binding in vitro intrinsic clearance did not separate the two groups of compounds, and in fact, paroxetine, a nonhepatotoxin, showed the greatest amount of covalent binding in microsomes. Including factors such as the fraction of total metabolism comprised by covalent binding and the total daily dose of each drug improved the discrimination between hepatotoxic and nontoxic drugs based on in vitro covalent binding data; however, the approach still would falsely identify some agents as potentially hepatotoxic.


Asunto(s)
Evaluación Preclínica de Medicamentos , Hepatocitos/efectos de los fármacos , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Pruebas de Toxicidad/métodos , Acetaminofén/química , Acetaminofén/metabolismo , Acetaminofén/farmacología , Sitios de Unión , Buspirona/química , Buspirona/metabolismo , Buspirona/farmacología , Carbamazepina/química , Carbamazepina/metabolismo , Carbamazepina/farmacología , Diclofenaco/química , Diclofenaco/metabolismo , Diclofenaco/farmacología , Difenhidramina/química , Difenhidramina/metabolismo , Difenhidramina/farmacología , Relación Dosis-Respuesta a Droga , Hepatocitos/metabolismo , Humanos , Indometacina/química , Indometacina/metabolismo , Indometacina/farmacología , Meloxicam , Microsomas Hepáticos/efectos de los fármacos , Estructura Molecular , Paroxetina/química , Paroxetina/metabolismo , Paroxetina/farmacología , Piperazinas , Propranolol/química , Propranolol/metabolismo , Propranolol/farmacología , Clorhidrato de Raloxifeno/química , Clorhidrato de Raloxifeno/metabolismo , Clorhidrato de Raloxifeno/farmacología , Simvastatina/química , Simvastatina/metabolismo , Simvastatina/farmacología , Relación Estructura-Actividad , Tiazinas/química , Tiazinas/metabolismo , Tiazinas/farmacología , Tiazoles/química , Tiazoles/metabolismo , Tiazoles/farmacología , Ticrinafeno/química , Ticrinafeno/metabolismo , Ticrinafeno/farmacología , Triazoles/química , Triazoles/metabolismo
7.
Chem Res Toxicol ; 20(12): 1954-65, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17935300

RESUMEN

The current study examined the bioactivation potential of a nonpeptidyl thrombopoietin receptor agonist, 1-(3-chloro-5-((4-(4-fluoro-3-(trifluoromethyl)phenyl)thiazol-2-yl)carbamoyl)pyridine-2-yl)piperidine-4-carboxylic acid (1), containing a 2-carboxamido-4-arylthiazole moiety in the core structure. Toxicological risks arising from P450-catalyzed C4-C5 thiazole ring opening in 1 via the epoxidation-->diol sequence were alleviated, since mass spectrometric analysis of human liver microsome and/or hepatocyte incubations of 1 did not reveal the formation of reactive acylthiourea and/or glyoxal metabolites, which are prototypic products derived from thiazole ring scission. However, 4-(4-fluoro-3-(trifluoromethyl)phenyl)thiazol-2-amine (2), the product of hydrolysis of 1 in human liver microsomes, hepatocytes, and plasma, underwent oxidative bioactivation in human liver microsomes, since trapping studies with glutathione led to the formation of two conjugates derived from the addition of the thiol nucleophile to 2 and a thiazole- S-oxide metabolite of 2. Mass spectral fragmentation and NMR analysis indicated that the site of attachment of the glutathionyl moiety in both conjugates was the C5 position in the thiazole ring. Based on the structures of the glutathione conjugates, two bioactivation pathways are proposed, one involving beta-elimination of an initially formed hydroxylamine metabolite and the other involving direct two-electron oxidation of the electron-rich 2-aminothiazole system to electrophilic intermediates. This mechanistic insight into the bioactivation process allowed the development of a rational chemical intervention strategy that involved blocking the C5 position with a fluorine atom or replacing the thiazole ring with a 1,2,4-thiadiazole group. These structural changes not only abrogated the bioactivation liability associated with 1 but also resulted in compounds that retained the attractive pharmacological and pharmacokinetic attributes of the prototype agent.


Asunto(s)
Piridinas/farmacología , Receptores de Trombopoyetina/agonistas , Tiazoles/química , Animales , Disponibilidad Biológica , Biotransformación , Línea Celular , Estabilidad de Medicamentos , Glutatión/metabolismo , Hepatocitos/metabolismo , Humanos , Masculino , Ratones , Microsomas Hepáticos/metabolismo , Estructura Molecular , Piridinas/sangre , Piridinas/química , Piridinas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Trombopoyetina/genética , Tiazoles/sangre , Tiazoles/metabolismo , Tiazoles/farmacología , Transfección
8.
Chem Res Toxicol ; 20(11): 1649-57, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17907785

RESUMEN

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.


Asunto(s)
Microsomas Hepáticos/metabolismo , NADP/metabolismo , Paroxetina/metabolismo , Quinonas/metabolismo , Biotransformación , Inhibidores del Citocromo P-450 CYP2D6 , Glutatión/metabolismo , Humanos , Quinonas/química , Tritio
9.
Drug Metab Dispos ; 35(11): 2111-8, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17686907

RESUMEN

The role of transporters in the disposition of (+)-2-[4-({[2-(benzo[1,3]dioxol-5-yloxy)-pyridine-3-carbonyl]-amino}-methyl)-3-fluoro-phenoxy]-propionic acid (CP-671,305), an orally active inhibitor of phosphodiesterase-4, was examined. In bile duct-exteriorized rats, a 7.4-fold decrease in the half-life of CP-671,305 was observed, implicating enterohepatic recirculation. Statistically significant differences in CP-671,305 pharmacokinetics (clearance and area under the curve) were discernible in cyclosporin A- or rifampicin-pretreated rats. Considering that cyclosporin A and rifampicin inhibit multiple uptake/efflux transporters, the interactions of CP-671,305 with major human hepatic drug transporters, multidrug resistance protein 1 (MDR1), multidrug resistance-associated protein 2 (MRP2), breast cancer resistant protein (BCRP), and organic anion-transporting polypeptide (OATPs) were evaluated in vitro. CP-671,305 was identified as a substrate of MRP2 and BCRP, but not MDR1. CP-671,305 was a substrate of human OATP2B1 with a high affinity (Km = 4 microM) but not a substrate for human OATP1B1 or OATP1B3. Consistent with these results, examination of hepatobiliary transport of CP-671,305 in hepatocytes indicated active uptake followed by efflux into bile canaliculi. Upon examination as a substrate for major rat hepatic Oatps, CP-671,305 displayed high affinity (Km = 12 microM) for Oatp1a4. The role of rat Mrp2 in the biliary excretion was also examined in Mrp2-deficient rats. The observations that CP-671,305 pharmacokinetics were largely unaltered suggested that compromised biliary clearance of CP-671,305 was compensated by increased urinary clearance. Overall, these studies suggest that hepatic transporters play an important role in the disposition and clearance of CP-671,305 in rat and human, and as such, these studies should aid in the design of clinical drug-drug interaction studies.


Asunto(s)
Proteínas de Transporte de Membrana/metabolismo , Inhibidores de Fosfodiesterasa 4 , Inhibidores de Fosfodiesterasa/farmacocinética , Propionatos/farmacocinética , Piridinas/farmacocinética , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Bilis/metabolismo , Células CHO , Línea Celular , Cricetinae , Cricetulus , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Masculino , Proteínas de Transporte de Membrana/genética , Estructura Molecular , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/genética , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/metabolismo , Propionatos/química , Propionatos/metabolismo , Piridinas/química , Piridinas/metabolismo , Ratas , Ratas Mutantes , Ratas Sprague-Dawley , Ratas Wistar , Transfección
10.
J Med Chem ; 49(14): 4425-36, 2006 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-16821801

RESUMEN

N-[(3R)-1-Azabicyclo[2.2.2]oct-3-yl]furo[2,3-c]pyridine-5-carboxamide (14, PHA-543,613), a novel agonist of the alpha7 neuronal nicotinic acetylcholine receptor (alpha7 nAChR), has been identified as a potential treatment of cognitive deficits in schizophrenia. Compound 14 is a potent and selective alpha7 nAChR agonist with an excellent in vitro profile. The compound is characterized by rapid brain penetration and high oral bioavailability in rat and demonstrates in vivo efficacy in auditory sensory gating and, in an in vivo model to assess cognitive performance, novel object recognition.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/síntesis química , Trastornos del Conocimiento/tratamiento farmacológico , Agonistas Nicotínicos/síntesis química , Nootrópicos/síntesis química , Quinuclidinas/síntesis química , Receptores Nicotínicos/metabolismo , Esquizofrenia/tratamiento farmacológico , Animales , Disponibilidad Biológica , Encéfalo/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Estabilidad de Medicamentos , Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Potenciales Evocados Auditivos/efectos de los fármacos , Humanos , Técnicas In Vitro , Aprendizaje/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología , Agonistas Nicotínicos/farmacocinética , Agonistas Nicotínicos/farmacología , Nootrópicos/farmacocinética , Nootrópicos/farmacología , Técnicas de Placa-Clamp , Quinuclidinas/química , Quinuclidinas/farmacología , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Receptores Nicotínicos/fisiología , Reconocimiento en Psicología/efectos de los fármacos , Estereoisomerismo , Relación Estructura-Actividad , Receptor Nicotínico de Acetilcolina alfa 7
11.
Chem Biol Interact ; 155(1-2): 10-20, 2005 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-15978881

RESUMEN

Therapy with the antidepressant trazodone has been associated with several cases of idiosyncratic hepatotoxicity. While the mechanism of hepatotoxicity remains unknown, it is possible that reactive metabolites of trazodone play a causative role. Studies were initiated to determine whether trazodone undergoes bioactivation in human liver microsomes to electrophilic intermediates. LC/MS/MS analysis of incubations containing trazodone and NADPH-supplemented microsomes or recombinant P4503A4 in the presence of glutathione revealed the formation of conjugates derived from the addition of the sulfydryl nucleophile to mono-hydroxylated- and hydrated-trazodone metabolites. Product ion spectra suggested that mono-hydroxylation and sulfydryl conjugation occurred on the 3-chlorophenyl-ring, whereas hydration and subsequent sulfydryl conjugation had occurred on the triazolopyridinone ring system. These findings are consistent with bioactivation sequences involving: (1) aromatic hydroxylation of the 3-chlorophenyl-ring in trazodone followed by the two-electron oxidation of this metabolite to a reactive quinone-imine intermediate, which reacts with glutathione in a 1,4-Michael fashion and (2) oxidation of the pyridinone ring to an electrophilic epoxide, ring opening of which, by glutathione or water generates the corresponding hydrated-trazodone-thiol conjugate or the stable diol metabolite, respectively. The pathway involving trazodone bioactivation to the quinone-imine has also been observed with many para-hydroxyanilines including the structurally related antidepressant nefazodone. It is proposed that the quinone-imine and/or the epoxide intermediate(s) may represent a rate-limiting step in the initiation of trazodone-mediated hepatotoxicity.


Asunto(s)
Antidepresivos de Segunda Generación/farmacocinética , Benzoquinonas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Compuestos Epoxi/metabolismo , Microsomas Hepáticos/metabolismo , Trazodona/farmacocinética , Biotransformación , Cromatografía Líquida de Alta Presión , Citocromo P-450 CYP3A , Humanos , Iminas/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Microsomas Hepáticos/efectos de los fármacos , Proteínas Recombinantes , Espectrometría de Masa por Ionización de Electrospray
12.
J Pharm Sci ; 94(1): 38-45, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15761928

RESUMEN

Higher-throughput ADME programs in early drug discovery are becoming common throughout the pharmaceutical industry as companies strive to reduce their compound attrition in later-stage development. Many of the ADME assays developed into higher-throughput formats rely on LC/MS analyses. Since the biological aspects of the assay are amenable to parallel processes using dense plate formats, the number of samples generated from these assays produce a large analysis load for serial LC/MS. Presented in this report are two novel strategies, including a sample pooling method and a two time-point method, that could be used in drug discovery to reduce the number of samples generated during multiple time-point in-vitro ADME assays. One hundred and sixty-three compounds were subjected to human microsomal incubations with full time-point method samples taken at t = 0, 5, 15, 30, and 45 min. The ER data correlation (R(2)) between the full time-point method and the pooling method and two time-point methods were 0.98 and 0.97, respectively. Both methods have the potential to: 1. produce data of similar quality to traditional high throughput ADME assays, 2. be easily implemented, 3. shorten analytical run times, and 4. be reproducible and robust.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Preparaciones Farmacéuticas/metabolismo , Algoritmos , Cromatografía Líquida de Alta Presión , Sistema Enzimático del Citocromo P-450/metabolismo , Interpretación Estadística de Datos , Semivida , Humanos , Cinética , Espectrometría de Masas , Microsomas Hepáticos/enzimología
13.
Drug Metab Dispos ; 33(2): 243-53, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15523046

RESUMEN

The therapeutic benefits of the antidepressant nefazodone have been hampered by several cases of acute hepatotoxicity/liver failure. Although the mechanism of hepatotoxicity remains unknown, it is possible that reactive metabolites of nefazodone play a causative role. Studies were initiated to determine whether nefazodone undergoes bioactivation in human liver microsomes to electrophilic intermediates. Following incubation of nefazodone with microsomes or recombinant P4503A4 in the presence of sulfydryl nucleophiles, conjugates derived from the addition of thiol to a monohydroxylated nefazodone metabolite were observed. Product ion spectra suggested that hydroxylation and sulfydryl conjugation occurred on the 3-chlorophenylpiperazine-ring, consistent with a bioactivation pathway involving initial formation of p-hydroxynefazodone, followed by its two-electron oxidation to the reactive quinone-imine intermediate. The formation of novel N-dearylated nefazodone metabolites was also discernible in these incubations, and 2-chloro-1,4-benzoquinone, a by-product of N-dearylation, was trapped with glutathione to afford the corresponding hydroquinone-sulfydryl adduct. Nefazodone also displayed NADPH-, time-, and concentration-dependent inactivation of P4503A4 activity, suggesting that reactive metabolites derived from nefazodone bioactivation are capable of covalently modifying P4503A4. A causative role for 2-chloro-1,4-benzoquinone and/or the quinone-imine intermediate(s) in nefazodone hepatotoxicity is speculated. Although the antianxiety agent buspirone, which contains a pyrimidine ring in place of the 3-chlorophenyl-ring, also generated p-hydroxybuspirone in liver microsomes, no sulfydryl conjugates of this metabolite were observed. This finding is consistent with the proposal that two-electron oxidation of p-hydroxybuspirone to the corresponding quinone-imine is less favorable due to differences in the protonation state at physiological pH and due to weaker resonance stabilization of the oxidation products as predicted from ab initio measurements.


Asunto(s)
Antidepresivos de Segunda Generación/metabolismo , Benzoquinonas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Triazoles/metabolismo , Antidepresivos de Segunda Generación/análisis , Antidepresivos de Segunda Generación/química , Benzoquinonas/análisis , Benzoquinonas/química , Biotransformación , Citocromo P-450 CYP3A , Inhibidores Enzimáticos del Citocromo P-450 , Humanos , Iminas/análisis , Iminas/química , Iminas/metabolismo , Piperazinas , Triazoles/análisis , Triazoles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...