Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Drug Metab Dispos ; 39(9): 1668-73, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21673127

RESUMEN

The novel biotransformation of an aminopyrrolidine to an aminopiperidine during the metabolism of 5-(4-chlorophenyl)-3-methyl-2-((2R)-2-(((1-methylethyl)amino)methyl)-1-pyrrolidinyl)-6-(4-pyridinyl)-4(3H)-pyrimidinone (AMG657417) was investigated using the NADPH-fortified S9 fraction from human liver. The major metabolite (M18) had a protonated molecule (MH(+) m/z 438) identical to that of AMG657417 except that it eluted earlier on a reverse-phase high-performance liquid chromatography. The structure of M18 had been identified as 5-(4-chlorophenyl)-3-methyl-2-((1-(1-methylethyl)-3-piperidinyl)amino)-6-(4-pyridinyl)-4(3H)-pyrimidinone (I) by liquid chromatography-mass spectrometry and proton NMR. M18 was not observed when AMG657417 was incubated with either microsomal or cytosolic fraction from human liver, suggesting the involvement of both microsomal and cytosolic enzymes in the biotransformation. The reaction mechanisms have been elucidated by trapping the intermediates formed during the biotransformation. An aldehyde intermediate was initially produced by hydroxylation and opening of the pyrrolidine ring of the parent molecule, followed by intramolecular Schiff-base formation between the exocyclic isopropylamine nitrogen and the aldehyde carbonyl to form a piperidinyl iminium ion. The iminium ion was then reduced to the piperidine product. The presence of the aldehyde intermediate was verified by the formation of semicarbazide conjugates in human liver microsomal, S9, and recombinant CYP3A4 incubations of AMG657417. The presence of the piperidinyl iminium ion intermediate was confirmed by the formation of cyanide conjugates in the incubations in human liver S9. Two cyanide conjugates with identical protonated molecule and product ion mass spectra were observed, indicating the likelihood of diastereomer formation. A chemical inhibition study in NADPH-fortified S9 fraction indicated that the oxidation of AMG657417 was catalyzed almost exclusively by CYP3A.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Piperidinas/farmacocinética , Pirimidinonas/farmacocinética , Pirrolidinas/farmacocinética , Biotransformación , Cromatografía Líquida de Alta Presión/métodos , Humanos , Hidroxilación , Cetoconazol/farmacología , Espectroscopía de Resonancia Magnética/métodos , Espectrometría de Masas/métodos , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Piperidinas/metabolismo , Pirimidinonas/metabolismo , Pirrolidinas/metabolismo , Semicarbacidas/metabolismo
2.
Drug Metab Dispos ; 39(7): 1139-48, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21441468

RESUMEN

Rifampin and carbamazepine have been recommended in the U.S. Food and Drug Administration draft drug interaction guidance as CYP3A4 inducers for clinical drug-drug interaction (DDI) studies. To optimize the dose regimens of these inducers for use in DDI studies, their effect at various doses and dosing durations on the area under the curve (AUC) of multiple probe substrates was simulated using a population-based simulator. A similar assessment of the inducer phenobarbital was also conducted. CYP3A4 induction by all three inducers was previously determined in hepatocytes, and the results were incorporated into simulations. The pharmacokinetics of the three inducers and their associated CYP3A4 drug interactions were predicted and compared with in vivo observations. The predicted C(max) and AUC of all the inducers and substrates correlated closely with those observed clinically. The predicted magnitudes of the DDIs caused by CYP3A4 induction were also in good agreement with the observed clinical results. Comparison of the maximal CYP3A4 induction potential among the three inducers indicated that rifampin is the most potent inducer and is the best choice for clinical CYP3A4 induction DDI studies. Moreover, a near-maximal CYP3A4 DDI was predicted to result from administration of rifampin for approximately 7 days at 450 to 600 mg q.d. or 200 to 300 mg b.i.d. These results suggest optimal dose regimens for clinical trials that maximize the probability of detecting a DDI caused by CYP3A4 induction. The simulation strategy provides the means to predict the induction profiles of compounds in development.


Asunto(s)
Citocromo P-450 CYP3A/biosíntesis , Interacciones Farmacológicas , Hepatocitos/enzimología , Área Bajo la Curva , Carbamazepina/farmacología , Inducción Enzimática , Humanos , Midazolam/farmacología , Nifedipino/farmacología , Fenobarbital/farmacología , Rifampin/farmacología , Simvastatina/farmacología , Especificidad por Sustrato
3.
Drug Metab Dispos ; 37(12): 2330-9, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19773538

RESUMEN

Time-dependent inactivation (TDI) of human cytochromes P450 3A4 (CYP3A4) is a major cause of clinical drug-drug interactions (DDIs). Human liver microsomes (HLM) are commonly used as an enzyme source for evaluating the inhibition of CYP3A4 by new chemical entities. The inhibition data can then be extrapolated to assess the risk of human DDIs. Using this approach, under- and overpredictions of in vivo DDIs have been observed. In the present study, human hepatocytes were used as an alternative to HLM. Hepatocytes incorporate the effects of other mechanisms of drug metabolism and disposition (i.e., phase II enzymes and transporters) that may modulate the effects of TDI on clinical DDIs. The in vitro potency (K(I) and k(inact)) of five known CYP3A4 TDI drugs (clarithromycin, diltiazem, erythromycin, verapamil, and troleandomycin) was determined in HLM (pooled, n = 20) and hepatocytes from two donors (D1 and D2), and the results were extrapolated to predict in vivo DDIs using a Simcyp population trial-based simulator. Compared with observed DDIs, the predictions derived from HLM appeared to be overestimated. The predictions based on TDI measured in hepatocytes were better correlated with the DDIs (n = 37) observed in vivo (R(2) = 0.601 for D1 and 0.740 for D2) than those from HLM (R(2) = 0.451). In addition, with the use of hepatocytes a greater proportion of the predictions were within a 2-fold range of the clinical DDIs compared with using HLM. These results suggest that DDI predictions from CYP3A4 TDI kinetics in hepatocytes could provide an alternative approach to balance HLM-based predictions that can sometimes substantially overestimate DDIs and possibly lead to erroneous conclusions about clinical risks.


Asunto(s)
Simulación por Computador , Inhibidores del Citocromo P-450 CYP3A , Inhibidores Enzimáticos/farmacología , Hepatocitos/efectos de los fármacos , Modelos Biológicos , Adulto , Anciano de 80 o más Años , Células Cultivadas , Citocromo P-450 CYP3A/metabolismo , Interacciones Farmacológicas , Femenino , Hepatocitos/enzimología , Humanos , Hidroxilación , Cinética , Masculino , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Midazolam/metabolismo , Persona de Mediana Edad , Reproducibilidad de los Resultados , Adulto Joven
4.
Rapid Commun Mass Spectrom ; 23(10): 1446-50, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19353558

RESUMEN

In vitro metabolism of imatinib was investigated in rat and human liver microsomes. Atmospheric pressure chemical ionization (APCI) mass spectrometry (MS) was applied in differentiating hydroxyl metabolites from N-oxides of imatinib because N-oxides are known to undergo deoxygenation during APCI. In addition, the major oxidative metabolite (M9, N-oxidation on the piperazine ring) was observed to undergo in-source fragmentation by elimination of formaldehyde. This fragment ion resulted from Meisenheimer rearrangement with migration of the N-methyl group to the corresponding N-methoxyl piperazine, followed by elimination of formaldehyde due to thermal energy activation at the vaporizer of APCI source. The presence of this fragment ion distinguished not only N-oxide from isomeric hydroxylated metabolite, but also unambiguously indicated that oxidation occurred on the N-4 of the piperazine ring where the methyl group was attached.


Asunto(s)
Cromatografía Liquida/métodos , Espectrometría de Masas/métodos , Microsomas Hepáticos/metabolismo , Piperazinas/metabolismo , Pirimidinas/metabolismo , Animales , Benzamidas , Humanos , Hidroxilación , Mesilato de Imatinib , Microsomas Hepáticos/química , Oxidación-Reducción , Piperazinas/química , Pirimidinas/química , Ratas
5.
Drug Metab Dispos ; 37(5): 925-31, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19196844

RESUMEN

Cytochrome P450 (P450) is the superfamily of enzymes responsible for biotransformation of endobiotics and xenobiotics. However, their large isoform multiplicity, inducibility, diverse structure, widespread distribution, polymorphic expression, and broad overlapping substrate specificity make it difficult to measure the precise role of each individual P450 to the metabolism of drugs (or carcinogens) and hamper the understanding of the relationship between the genetic/environmental factors that regulate P450 phenotype and the responses of the individual P450s to drugs. The antibodies against P450s have been useful tools for the quantitative determination of expression level and contribution of the epitope-specific P450 to the metabolism of a drug or carcinogen substrate in tissues containing multiple P450 isoforms and for implications in pharmacogenetics and human risk assessment. In particular, the inhibitory antibodies are uniquely suited for reaction phenotyping that helps to predict human pharmacokinetics for clinical drug-drug interaction potential in drug discovery and development.


Asunto(s)
Sistema Enzimático del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/inmunología , Animales , Anticuerpos Bloqueadores/química , Anticuerpos Bloqueadores/farmacología , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Isoenzimas/metabolismo , Fenotipo , Medición de Riesgo
6.
Pharm Res ; 26(2): 459-68, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19082692

RESUMEN

PURPOSE: Gaboxadol, a selective extrasynaptic agonist of the delta-containing gamma-aminobutyric acid type A (GABAA) receptor, is excreted in humans into the urine as parent drug and glucuronide conjugate. The goal of this study was to identify the UDP-Glucuronosyltransferase (UGT) enzymes and the transporters involved in the metabolism and active renal secretion of gaboxadol and its metabolite in humans.Methods. The structure of the glucuronide conjugate of gaboxadol in human urine was identified by LC/MS/MS. Human recombinant UGT isoforms were used to identify the enzymes responsible for the glucuronidation of gaboxadol. Transport of gaboxadol and its glucuronide was evaluated using cell lines and membrane vesicles expressing human organic anion transporters hOAT1 and hOAT3, organic cation transporter hOCT2, and the multidrug resistance proteins MRP2 and MRP4.Results. Our study indicated that the gaboxadol-O-glucuronide was the major metabolite excreted in human urine. UGT1A9, and to a lesser extent UGT1A6, UGT1A7 and UGT1A8, catalyzed the O-glucuronidation of gaboxadol in vitro. Gaboxadol was transported by hOAT1, but not by hOCT2, hOAT3, MRP2, and MRP4. Gaboxadol-O-glucuronide was transported by MRP4, but not MRP2.Conlusion. Gaboxadol could be taken up into the kidney by hOAT1 followed by glucuronidation and efflux of the conjugate into urine via MRP4.


Asunto(s)
Agonistas del GABA/farmacocinética , Glucurónidos/metabolismo , Glucuronosiltransferasa/metabolismo , Isoxazoles/farmacocinética , Riñón/enzimología , Hígado/enzimología , Proteínas de Transporte de Membrana/metabolismo , Administración Oral , Animales , Biotransformación , Células CHO , Cromatografía Liquida , Cricetinae , Cricetulus , Agonistas del GABA/administración & dosificación , Agonistas del GABA/orina , Glucuronosiltransferasa/genética , Humanos , Isoenzimas , Isoxazoles/administración & dosificación , Isoxazoles/orina , Proteínas de Transporte de Membrana/genética , Microsomas Hepáticos/enzimología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Proteínas Recombinantes/metabolismo , Espectrometría de Masas en Tándem , Transfección , UDP Glucuronosiltransferasa 1A9
7.
Drug Metab Dispos ; 36(12): 2414-8, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18799804

RESUMEN

Imatinib, a potent and selective protein tyrosine kinase inhibitor, has been approved for the treatment of chronic myelogenous leukemia and metastatic and unresectable malignant gastrointestinal stromal tumors. In vitro metabolism of imatinib was investigated in rat and human liver microsomes. Besides several oxidative metabolites and an N-desmethyl metabolite, as previous reported, a novel metabolite with a mass addition of 621 atomic mass units to the parent was detected as the major metabolite in the incubations with rat liver microsomes, using NADPH as a cofactor. The analysis of MS(2) and MS(n) data revealed that this metabolite corresponded to adenine dinucleotide phosphate (ADP+) conjugate of imatinib. The ADP+ adduct was scaled up from rat liver microsomal incubations and isolated for NMR analysis. NMR data confirmed and conclusively showed the conjugation had occurred between the pyridine nitrogen of imatinib to the ribose ring of ADP+ moiety. The formation of this adduct was enzymatic and required NADP+ as a reactant. In addition, ADP+ adducts of imatinib N-oxide and desmethyl imatinib were also detected as minor metabolites in the incubations with rat liver microsomes. In contrast, only trace levels of ADP+ adducts of imatinib and desmethyl imatinib were detected in the incubations with human liver microsomes. Imatinib-ADP+ adducts have been observed only in in vitro studies to date. The physiological role of these adducts is not clear, nor is their in vivo relevance.


Asunto(s)
Microsomas Hepáticos/metabolismo , NADP/análogos & derivados , NADP/metabolismo , Piperazinas/metabolismo , Pirimidinas/metabolismo , Animales , Benzamidas , Cromatografía Líquida de Alta Presión , Humanos , Mesilato de Imatinib , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Estructura Molecular , NAD+ Nucleosidasa/química , NADP/química , Piperazinas/química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Pirimidinas/química , Ratas
8.
Drug Metab Dispos ; 36(11): 2355-70, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18669588

RESUMEN

CYP3A4 induction is not generally considered to be a concern for safety; however, serious therapeutic failures can occur with drugs whose exposure is lower as a result of more rapid metabolic clearance due to induction. Despite the potential therapeutic consequences of induction, little progress has been made in quantitative predictions of CYP3A4 induction-mediated drug-drug interactions (DDIs) from in vitro data. In the present study, predictive models have been developed to facilitate extrapolation of CYP3A4 induction measured in vitro to human clinical DDIs. The following parameters were incorporated into the DDI predictions: 1) EC(50) and E(max) of CYP3A4 induction in primary hepatocytes; 2) fractions unbound of the inducers in human plasma (f(u, p)) and hepatocytes (f(u, hept)); 3) relevant clinical in vivo concentrations of the inducers ([Ind](max, ss)); and 4) fractions of the victim drugs cleared by CYP3A4 (f(m, CYP3A4)). The values for [Ind](max, ss) and f(m, CYP3A4) were obtained from clinical reports of CYP3A4 induction and inhibition, respectively. Exposure differences of the affected drugs in the presence and absence of the six individual inducers (bosentan, carbamazepine, dexamethasone, efavirenz, phenobarbital, and rifampicin) were predicted from the in vitro data and then correlated with those reported clinically (n = 103). The best correlation was observed (R(2) = 0.624 and 0.578 from two hepatocyte donors) when f(u, p) and f(u, hept) were included in the predictions. Factors that could cause over- or underpredictions (potential outliers) of the DDIs were also analyzed. Collectively, these predictive models could add value to the assessment of risks associated with CYP3A4 induction-based DDIs by enabling their determination in the early stages of drug development.


Asunto(s)
Citocromo P-450 CYP3A/biosíntesis , Modelos Biológicos , Modelos Químicos , Adulto , Alquinos , Benzoxazinas/farmacología , Bosentán , Carbamazepina/farmacología , Células Cultivadas , Ciclopropanos , Citocromo P-450 CYP3A/sangre , Citocromo P-450 CYP3A/química , Citocromo P-450 CYP3A/genética , Relación Dosis-Respuesta a Droga , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Femenino , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Humanos , Valor Predictivo de las Pruebas , Especificidad por Sustrato/efectos de los fármacos , Sulfonamidas/farmacología
9.
Curr Drug Metab ; 9(5): 384-94, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18537575

RESUMEN

In vivo enzyme levels are governed by the rates of de novo enzyme synthesis and degradation. A current lack of consensus on values of the in vivo turnover half-lives of human cytochrome P450 (CYP) enzymes places a significant limitation on the accurate prediction of changes in drug concentration-time profiles associated with interactions involving enzyme induction and mechanism (time)-based inhibition (MBI). In the case of MBI, the full extent of inhibition is also sensitive to values of enzyme turnover half-life. We review current understanding of CYP regulation, discuss the pros and cons of various in vitro and in vivo approaches used to estimate the turnover of specific CYPs and, by simulation, consider the impact of variability in estimates of CYP turnover on the prediction of enzyme induction and MBI in vivo. In the absence of consensus on values for the in vivo turnover half-lives of key CYPs, a sensitivity analysis of predictions of the pharmacokinetic effects of enzyme induction and MBI to these values should be an integral part of the modelling exercise, and the selective use of values should be avoided.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Animales , Sistema Enzimático del Citocromo P-450/biosíntesis , Inducción Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/genética , Regulación Enzimológica de la Expresión Génica/fisiología , Humanos , Mucosa Intestinal/metabolismo , Intestinos/enzimología , Cinética , Hígado/enzimología , Hígado/metabolismo
10.
Mol Pharm ; 5(3): 438-48, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18345638

RESUMEN

Many pharmacokinetic analyses require unbound plasma concentrations, including prediction of clearance, volume of distribution, drug-drug interactions, brain uptake analysis, etc. It is most often more convenient to measure the total drug concentration in plasma rather than the unbound drug concentration. To arrive at unbound plasma concentrations, separate in vitro determinations of the plasma protein binding of a drug are usually carried out in serum or in plasma, and the plasma pharmacokinetic results are then mathematically adjusted by this fraction unbound ( f u,p). Plasma protein binding or the drug fraction unbound in plasma ( f u,p) is known to be affected by protein, drug, free fatty acid concentrations, lipoprotein partitioning, temperature, pH, and the presence or absence of other drugs/displacing agents within plasma samples. Errors in f u,p determination caused by lack of adequate pH control in newer assay formats for plasma protein binding (e.g., 96-well equilibrium thin walled polypropylene dialysis plates) will have significant drug-specific impact on these pharmacokinetic calculations. Using a diverse set of 55 drugs and a 96-well equilibrium dialysis plate format, the effect of variable pH during equilibrium dialysis experiments on measured values of f u,p was examined. Equilibrium dialysis of human plasma against Dulbecco's phosphate buffered saline at 37 degrees C under an air or 10% CO 2 atmosphere for 22 h resulted in a final pH of approximately 8.7 and 7.4, respectively. The ratio of f u,p at pH 7.4 (10% CO 2) vs pH 8.7 (air) was >or=2.0 for 40% of the 55 compounds tested. Only one of the 55 compounds tested had a ratio <0.9. Select compounds were further examined in rat and dog plasma. In addition, physicochemical properties were calculated for all compounds using ACD/Labs software or Merck in-house software and compared to plasma protein binding results. Changes in plasma protein binding due to pH increases which occurred during the equilibrium dialysis experiment were not species specific but were drug-specific, though nonpolar, cationic compounds had a higher likely hood of displaying pH-dependent binding. These studies underscore the importance of effectively controlling pH in plasma protein binding studies.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Animales , Fenómenos Químicos , Química Física , Diálisis , Perros , Interacciones Farmacológicas , Femenino , Humanos , Concentración de Iones de Hidrógeno , Cinética , Plasma , Presión , Unión Proteica , Ratas , Especificidad de la Especie
11.
Drug Metab Dispos ; 34(12): 2073-8, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16959878

RESUMEN

Drug transporters have been shown to alter drug metabolism. Similarly, bioactivation of drugs may also be altered by drug transporters. The aim of this work was to examine the role of P-glycoprotein (Pgp) in the bioactivation of a Pgp substrate, raloxifene, and a non-Pgp substrate, naphthalene. To evaluate the extent of bioactivation, covalent binding was measured. In both freshly isolated and cryopreserved hepatocytes, the extent of raloxifene covalent binding increased significantly (p < 0.05) in the presence of verapamil, whereas no change was observed with the covalent binding of naphthalene. To ascertain that the change was a Pgp effect, covalent binding was examined in microsomes in which raloxifene and naphthalene covalent binding was not altered in the presence of verapamil. In addition, the measure of raloxifene-glutathione adducts in the cryopreserved hepatocytes showed that the formation of the adducts increased in the presence of verapamil, which supports the idea that blocking Pgp in the liver increases metabolism and, therefore, the bioactivation of raloxifene. Because raloxifene and naphthalene are known to undergo bioactivation mediated by CYP3A4, covalent binding in the presence of ketoconazole was examined. In both hepatocytes and microsomes, raloxifene covalent binding decreased significantly (p < 0.01). It is interesting that naphthalene covalent binding was not affected. In the presence of the CYP2E inhibitor 4-methylpyrazole, a decrease in naphthalene covalent binding was observed, suggesting that the formation of the 1,2-epoxide may be the main culprit contributing to naphthalene covalent binding. In conclusion, these data suggest that in addition to other "protective" mechanisms, Pgp may attenuate bioactivation of drugs.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Naftalenos/metabolismo , Clorhidrato de Raloxifeno/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Biotransformación , Células Cultivadas , Inhibidores del Citocromo P-450 CYP3A , Inhibidores Enzimáticos/farmacología , Fomepizol , Glutatión/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Cetoconazol/farmacología , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Pirazoles/farmacología , Verapamilo/farmacología
12.
Drug Metab Dispos ; 34(10): 1703-12, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16815964

RESUMEN

The rhesus monkey (Macaca mulatta) is a primate species used extensively as a preclinical safety species in drug development. In this report, we describe the cloning, expression, and characterization of CYP3A64 (AY334551), a CYP3A4 homolog expressed in rhesus liver. The deduced amino acid sequence was found to be 93% similar to human CYP3A4, 83% similar to human CYP3A5, and identical to the previously reported cynomolgus monkey CYP3A8 (Komori et al., 1992). The substrate specificity of CYP3A64 for testosterone (0-250 microM), midazolam (0-200 microM), nifedipine (0-200 microM), and 7-benzoxy-4-trifluoromethylcoumarin (0-200 microM) were compared with recombinant enzymes from rat (CYP3A1, CYP3A2), dog (CYP3A12, CYP3A26), rabbit (CYP3A6), and human (CYP3A4, CYP3A5). Immunoinhibition and chemical inhibition of CYP3A64 was demonstrated using the inhibitory monoclonal antibody (MAb) 10-1-1 (anti-3A4) and ketoconazole (0-10 microM). The utility of CYP3A64 to be used as a standard in monkey induction assays was shown and the concentration of CYP3A64 protein in rhesus liver microsomes was estimated to be 72 pmol/mg protein. In summary, these results support the utilization of rhesus monkey CYP3A64 for in vitro drug metabolism studies and provide a more complete understanding of CYP3A substrate specificities and species differences in metabolic capabilities.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Macaca mulatta/genética , Proteínas Recombinantes/metabolismo , Secuencia de Aminoácidos , Animales , Baculoviridae/genética , Secuencia de Bases , Western Blotting , Línea Celular , Clonación Molecular , Cumarinas/metabolismo , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/genética , ADN Complementario/química , ADN Complementario/genética , Humanos , Cetoconazol/metabolismo , Cinética , Hígado/enzimología , Hígado/metabolismo , Macaca mulatta/metabolismo , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Midazolam/metabolismo , Datos de Secuencia Molecular , Nifedipino/metabolismo , Proteínas Recombinantes/genética , Análisis de Secuencia de ADN , Spodoptera , Especificidad por Sustrato , Testosterona/metabolismo
13.
Eur J Pharm Sci ; 26(2): 151-61, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16085400

RESUMEN

Enterohepatic recirculation (EHR) occurs via biliary excretion and intestinal reabsorption of a drug. Drug recycling through EHR can lead to a change in pharmacokinetic (PK) properties, such as reduced clearance (CL), extended half-life (T(1/2)) and increased plasma exposure (AUC). As a result, EHR may prolong the pharmacological effect of drugs. In the present study, the compound (Cpd A) was found to exhibit EHR in Rhesus monkeys associated with a reduction in CL (from 3.8 to 0.33 Lh(-1), IV; from 2.3 to 0.4 Lh(-1), PO), and an increase in T(1/2) (from 0.9 to 18 h, IV) and in AUC (from 1.5 to 17.4 microg h/mL, IV; from 2.8 to 16.3 microg h/mL, PO), by comparing the PK in the monkeys via the interruption of EHR (bile-duct cannulation) with that in the intact monkeys. A population four-compartment model was constructed based on recirculation loops incorporating all possible inputs (bile secretion, a lag-time model for gall bladder emptying, routes and amounts of a single dose administration) to fully evaluate the EHR of Cpd A. The plasma concentrations versus time profiles predicted from the model had a good fit to the values observed in the subjects and were further simulated with 90% confidence interval to demonstrate its utility. Thus, the model could be applied as a useful tool to evaluate the drugs or compounds that undergo EHR in different species.


Asunto(s)
Bilis/metabolismo , Circulación Enterohepática , Modelos Biológicos , Farmacocinética , Animales , Área Bajo la Curva , Bilis/química , Biotransformación , Conducto Colédoco/cirugía , Semivida , Absorción Intestinal , Hígado/metabolismo , Macaca mulatta , Masculino , Tasa de Depuración Metabólica , Xenobióticos/farmacocinética , Xenobióticos/farmacología
14.
Curr Opin Drug Discov Devel ; 8(1): 66-77, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15679174

RESUMEN

There is great interest within the pharmaceutical industry in predicting the in vivo pharmacokinetics (PKs) and metabolism-based drug-drug interactions (DDIs) of compounds from their in vitro metabolism data. Metabolism-based DDIs are largely due to changes in levels of drug-metabolizing enzymes caused by one drug, leading to changes in the PK parameters (mainly clearance) of another. The search for alternative approaches to time-consuming and costly clinical PK drug interaction studies for predicting human DDIs, has been ongoing for decades. In vitro enzyme-mediated biotransformation reactions provide a foundation for predictions that relate PK concepts to enzyme kinetics. This review discusses the principles, assumptions, tools and approaches to in vitro/in vivo prediction, especially in the context of hepatic clearance (the most important PK parameter) and its prediction from in vitro data. Enzyme inhibition is a common cause of DDIs and involves various mechanisms (eg, reversible and mechanism-based inhibition). The models and equations used for predicting DDIs for different types of inhibitor (ie, competitive, partial competitive, non-competitive, partial non-competitive and mixed-type reversible inhibitors, and mechanism-based inhibitors) are extensively presented. Although the methods of prediction are numerous, there remain a number of unresolved factors that may affect the accuracy of the prediction. These factors are also discussed to provide a caution to researchers performing prediction studies.


Asunto(s)
Interacciones Farmacológicas , Preparaciones Farmacéuticas/metabolismo , Farmacocinética , Algoritmos , Animales , Disponibilidad Biológica , Inhibidores Enzimáticos/farmacología , Humanos
15.
J Pharmacol Exp Ther ; 313(2): 518-28, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15677349

RESUMEN

In the present study, N-(alpha-methylbenzyl-)-1-aminobenzotriazole (MBA) and ketoconazole (KET) were identified as the inhibitors with selectivity toward dog CYP2B11 and CYP3A12, respectively. Their selectivity was evaluated using phenacetin O-deethylation (CYP1A), diazepam (DZ) N1-demethylation (CYP2B11), diclofenac 4'-hydrxylation (CYP2C21), bufuralol 1'-hydroxylation (CYP2D11), and DZ C3-hydroxylation (CYP3A12) activities in dog liver microsomes (DLM). MBA exhibited potent mechanism-based inhibition of DZ N1-demethylase activity catalyzed by both baculovirus-expressed CYP2B11 and DLM. In both cases, inhibition was characterized by a low K(I) (0.35 and 0.46 microM, respectively) and high k(inact) (1.5 and 0.56 min(-1), respectively). Despite complete loss of DZ N1-demethylase activity in the presence of MBA, there was no significant loss of cytochrome P450 (P450) CO-binding spectrum. These data suggest that the inactivation involved covalent modification of P450 apoprotein, instead of the prosthetic heme moiety. A homology model of CYP2B11 was constructed, based on the crystal structure of rabbit CYP2C5, for docking the substrate (DZ) and the inhibitor (MBA), respectively. The model, within the limits of our approximations, helped explain the substrate specificity and inhibitor selectivity of CYP2B11. In contrast to MBA, KET was identified as a potent and selective reversible (competitive) inhibitor of CYP3A12 (K(I) = 0.13-0.33 microM). In fact, complete inhibition of CYP3A12-dependent DZ C3-hydroxylation was possible at a low KET concentration (1 microM). Therefore, it is concluded that one can attempt to conduct P450 reaction phenotype studies with DLM using MBA and KET as selective inhibitors of CYP2B11 and CYP3A12, respectively.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/antagonistas & inhibidores , Inhibidores Enzimáticos del Citocromo P-450 , Hígado/enzimología , Esteroide Hidroxilasas/antagonistas & inhibidores , Animales , Hidrocarburo de Aril Hidroxilasas/metabolismo , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Sistema Enzimático del Citocromo P-450/metabolismo , Familia 2 del Citocromo P450 , Perros , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Cetoconazol/farmacología , Hígado/efectos de los fármacos , Esteroide Hidroxilasas/metabolismo
16.
Drug Metab Dispos ; 32(11): 1299-303, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15483196

RESUMEN

Sulfation of ethinyl estradiol (EE) is a major pathway of first pass metabolism in both the intestine and liver. Consequently, we sought to identify the human sulfotransferases (SULTs) involved in the 3-O-sulfation of EE (EE-SULT). Based on the results described herein, cDNA-expressed human cytosolic SULT1A3 and SULT1E1 were identified as low Km isoforms (18.9 and 6.7 nM, respectively) mediating the sulfation of EE. In contrast, the EE-SULT catalyzed by other recombinant SULTs (SULT1A1 and 2A1) was a relatively high Km process (Km > or = 230 nM). The kinetics of EE-SULT in human intestine (Km1 = 24 nM; Km2 = 1206 nM) and liver (Km1 = 8 nM; Km2 = 2407 nM) cytosol was biphasic and conformed to a two-Km model with both low and high Km components. At a low EE concentration (3 nM), inhibition of EE-SULT activity (intestinal) was characterized with 2,6-dichloro p-nitrophenol (DCNP) (IC50 = 15.6 microM) and quercetin (IC50 = 0.4 microM). When these IC50 values were compared with those derived from expressed enzyme, inhibition of EE-SULT was consistent with the SULT1E1 (DCNP, IC50 = 20 microM; quercetin, IC50 = 0.6 microM), but not SULT1A3 (DCNP, IC50 = 12.4; quercetin, IC50 = 7 microM). Moreover, when estrone (which selectively inhibits expressed SULT1E1 and SULT1A3) was included in intestinal incubations, the high-affinity component of the Eadie-Hofstee plot for EE sulfation was inhibited, converting the plot from biphasic to monophasic. Collectively, these data are consistent with SULT1E1 as the primary sulfotransferase involved in EE sulfation at clinically relevant concentrations (<10 nM).


Asunto(s)
Etinilestradiol/metabolismo , Sulfotransferasas/metabolismo , Humanos , Isoenzimas/metabolismo , Sulfatos/metabolismo
18.
Drug Metab Dispos ; 32(8): 848-61, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15258111

RESUMEN

Compound A (3-[2-oxo-3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyrindin-2-yl)propyl]-imidazolidin-1-yl]-3(S)-(6-methoxy-pyridin-3-yl)-propionic acid), a potent and selective antagonist of integrin alpha(v)beta(3) receptor, is under development for treatment of osteoporosis. This study describes metabolism and excretion of A in vivo in rats, dogs, and monkeys, and metabolism of A in vitro in primary hepatocytes from rats, dogs, monkeys, and humans. In all three animal species studied, A was primarily excreted as unchanged drug and, to a lesser degree, as phase I and phase II metabolites. Major biotransformation pathways of A included glucuronidation/glucosylation on the carboxylic group to form acyl-linked glucuronides/glucosides; and oxidation on the tetrahydronaphthyridine moiety to generate a carbinolamine and its further metabolized products. Minor pathways involved O-demethylation and hydroxylations on the alkyl chain. Only in rats, a glutathione adduct of A was also observed, and its formation is proposed to be via an iminium intermediate on the tetrahydronaphthyridine ring. Similar metabolic pathways were observed in the incubates of hepatocytes from the corresponding animals as well as from humans. CYP 3A and 2D subfamilies were capable of metabolizing A to its oxidative products. Overall, these in vitro and in vivo findings should provide useful insight on possible biotransformation pathways of A in humans.


Asunto(s)
Integrina alfaVbeta3/antagonistas & inhibidores , Integrina alfaVbeta3/metabolismo , Animales , Perros , Femenino , Integrina alfaVbeta3/análisis , Macaca mulatta , Masculino , Ratas , Ratas Sprague-Dawley
19.
Drug Metab Dispos ; 32(6): 584-6, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15155548

RESUMEN

(-)-N-3-Benzyl-phenobarbital (NBPB) was reported to be a potent and selective inhibitor of CYP2C19. To validate the selectivity of NBPB toward CYP2C19 in human liver microsomes, the inhibitory effects on major cytochrome P450 isoform-specific reactions were evaluated in the present study. In human liver microsomes, NBPB showed potent competitive inhibition on CYP2C19-mediated S-mephenytoin 4'-hydroxylation with an IC(50) value of 0.25 microM and K(i) value of 0.12 microM, whereas weak inhibition was observed for CYP1A2-, CYP2A6-, CYP2B6-, CYP2C8-, CYP2C9-, CYP2D6-, and CYP3A4-mediated reactions with IC(50) values >100, >100, 62, 34, 19, >100, and 89 microM, respectively. Importantly, its selectivity toward CYP2C19 among the CYP2C subfamily was demonstrated. Therefore, NBPB can be used as a potent and selective inhibitor to establish the relative contribution of CYP2C19 for in vitro reaction phenotyping studies. This compound can also serve as a positive control inhibitor of CYP2C19 for routine screening of P450 reversible inhibition when human liver microsomes are used as the enzyme source.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/antagonistas & inhibidores , Microsomas Hepáticos/efectos de los fármacos , Oxigenasas de Función Mixta/antagonistas & inhibidores , Fenobarbital/análogos & derivados , Fenobarbital/farmacología , Cromatografía Líquida de Alta Presión , Citocromo P-450 CYP2C19 , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Técnicas In Vitro , Masculino , Espectrometría de Masas , Microsomas Hepáticos/enzimología , Fenobarbital/síntesis química
20.
Biochemistry ; 42(48): 14284-92, 2003 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-14640697

RESUMEN

The known activity of cytochrome P450 46A1 (P450 46A1) is 24(S)-hydroxylation of cholesterol. This reaction produces biologically active oxysterol, 24(S)-hydroxycholesterol, and is also the first step in enzymatic degradation of cholesterol in the brain. We report here that P450 46A1 can further metabolize 24(S)-hydroxycholesterol, giving 24,25- and 24,27-dihydroxycholesterols in both the cell cultures transfected with P450 46A1 cDNA and the in vitro reconstituted system with recombinant enzyme. In addition, P450 46A1 was able to carry out side chain hydroxylations of two endogenous C27-steroids with and without a double bond between C5-C6 (7alpha-hydroxycholesterol and cholestanol, respectively) and introduce a hydroxyl group on the steroid nucleus of the C21-steroid hormones with the C4-C5 double bond (progesterone and testosterone). Also, P450 46A1 was found to metabolize xenobiotics carrying out dextromethorphan O- and N-demethylations, diclofenac 4'-hydroxylation, and phenacetin O-deethylation. Thus, substrate specificities of P450 46A1 are not limited to cholesterol and include a number of structurally diverse compounds. Activities of P450 46A1 suggest that, in addition to the involvement in cholesterol homeostasis in the brain, this enzyme may participate in metabolism of neurosteroids and drugs that can cross the blood-brain barrier and are targeted to the central nervous system.


Asunto(s)
Encéfalo/enzimología , Colesterol/metabolismo , Esteroide Hidroxilasas/química , Esteroide Hidroxilasas/metabolismo , Sitios de Unión , Biotransformación , Encéfalo/metabolismo , Catálisis , Línea Celular , Colesterol 24-Hidroxilasa , Humanos , Hidroxicolesteroles/metabolismo , Hidroxilación , Preparaciones Farmacéuticas/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Esteroide Hidroxilasas/genética , Esteroide Hidroxilasas/aislamiento & purificación , Especificidad por Sustrato , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA