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










Base de datos
Intervalo de año de publicación
1.
Rapid Commun Mass Spectrom ; 22(21): 3434-42, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18837479

RESUMEN

A robust bioanalytical method capable of measuring acetyl and palmitoyl carnitines was developed and validated. Application of hydrophilic interaction chromatography (HILIC) enabled retention of these highly polar and difficult to analyze compounds on a silica HPLC column. The chromatography was conducted with a high percentage of an organic component in the mobile phase, allowing high sensitivity for the pre-existing positively charged quaternary ammonium ions by electrospray ionization mass spectrometry. Successful application of the method to reliably quantify naturally occurring acyl carnitines in mouse plasma depended on the use of corresponding deuterated analogues. The specificity of the method, achieved through the use of stable isotope labeled compounds in combination with a mass spectral multiple reaction monitoring technique, permitted a non-invasive assessment of the overall change in the levels of these acyl carnitines in the plasma of intact animals administered peroxisome proliferator activated receptor (PPAR) agents. These acyl carnitines, as carriers of the corresponding long-chain fatty acids for transport into mitochondria, can be employed as potential biomarkers for significant alteration in the beta-oxidation process in an intact animal.


Asunto(s)
Carnitina/análogos & derivados , Cromatografía Liquida/métodos , Espectrometría de Masas/métodos , Palmitoilcarnitina/sangre , Animales , Biomarcadores/sangre , Biomarcadores/química , Calibración , Carnitina/sangre , Marcaje Isotópico , Masculino , Ratones , Ratones Endogámicos , Estructura Molecular , Oxidación-Reducción , Palmitoilcarnitina/química , Estándares de Referencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
2.
Chem Res Toxicol ; 21(9): 1672-89, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18702535

RESUMEN

Stable isotope-labeled compounds have been synthesized and utilized by scientists from various areas of biomedical research during the last several decades. Compounds labeled with stable isotopes, such as deuterium and carbon-13, have been used effectively by drug metabolism scientists and toxicologists to gain better understanding of drugs' disposition and their potential role in target organ toxicities. The combination of stable isotope-labeling techniques with mass spectrometry and nuclear magnetic resonance (NMR) spectroscopy, which allows rapid acquisition and interpretation of data, has promoted greater use of these stable isotope-labeled compounds in absorption, distribution, metabolism, and excretion (ADME) studies. Examples of the use of stable isotope-labeled compounds in elucidating structures of metabolites and delineating complex metabolic pathways are presented in this review. The application of labeled compounds in mechanistic toxicity studies will be discussed by providing an example of how strategic placement of a deuterium atom in a drug molecule mitigated specific-specific renal toxicity. Other examples from the literature demonstrating the application of stable isotope-labeled compounds in understanding metabolism-mediated toxicities are presented. Furthermore, an example of how a stable isotope-labeled compound was utilized to better understand some of the gene changes in toxicogenomic studies is discussed. The interpretation of large sets of data produced from toxicogenomics studies can be a challenge. One approach that could be used to simplify interpretation of the data, especially from studies designed to link gene changes with the formation of reactive metabolites thought to be responsible for toxicities, is through the use of stable isotope-labeled compounds. This is a relatively unexplored territory and needs to be further investigated. The employment of analytical techniques, especially mass spectrometry and NMR, used in conjunction with stable isotope-labeled compounds to establish and understand mechanistic link between reactive metabolite formation, genomic, and proteomic changes and onset of toxicity is proposed. The use of stable isotope-labeled compounds in early human ADME studies as a way of identifying and possibly quantifying all drug-related components present in systemic circulation is suggested.


Asunto(s)
Alquinos/metabolismo , Bencilaminas/metabolismo , Bencilaminas/toxicidad , Ácido Glutámico/metabolismo , Animales , Bencilaminas/química , Ácido Glutámico/química , Humanos , Marcaje Isotópico
3.
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
4.
Chem Res Toxicol ; 20(10): 1503-12, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17900172

RESUMEN

Treatment with flutamide has been associated with clinical hepatotoxicty. The toxicity, metabolism,and transport of flutamide were investigated using cultured human hepatocytes. Flutamide and its major metabolite, 2-hydroxyflutamide, caused an inhibition of taurocholate efflux in human hepatocytes with an IC50=75 microM and 110 microM, respectively. Treatment of hepatocytes with flutamide or 2-hydroxyflutamide for 24 h resulted in time- and concentration-dependent toxicity as assessed by inhibition of protein synthesis. Toxicity was greater after 1 h than after 24 h of treatment. Recovery in inhibition of protein synthesis by 24 h was attributed to the decreased presence of flutamide due to its metabolism. Flutamide was metabolized by hepatocytes to several metabolites, and formation of reactive intermediates of flutamide, as evidenced by the presence of glutathione-related adducts, was observed. Inhibition of flutamide metabolism by 1-aminobenzotriazole (ABT) resulted in enhancement of flutamide toxicity, which was associated with sustained levels of nonmetabolized drug. ABT also prevented the formation of reactive intermediates of flutamide. There was an additive toxicity when cells were treated with a combination of flutamide and 2-hydroxyflutamide. Simultaneous treatment with flutamide and acetaminophen (APAP) resulted in additive to synergistic toxic effects. Flutamide and APAP were found to have significant effects on each other's metabolism. Flutamide inhibited glucuronidation and sulfation of APAP, resulting in greater amounts of APAP available for bioactivation. APAP inhibited the hydroxylation of flutamide, and subsequent sulfation and acetylation of 4-nitro-3-(trifluoromethyl) aniline, a metabolite of flutamide. In summary, we suggest that inhibition of bile acid efflux by flutamide and its 2-hydroxy metabolite may play a role in flutamide-induced liver injury. Both flutamide and 2-hydroxyflutamide are responsible for cytotoxicity if not metabolized. The data also suggest a possible drug-drug interaction between flutamide and APAP, resulting in inhibition of flutamide metabolism and increased APAP bioactivation and toxicity.


Asunto(s)
Acetaminofén/toxicidad , Analgésicos no Narcóticos/toxicidad , Antagonistas de Andrógenos/toxicidad , Flutamida/toxicidad , Hepatocitos/efectos de los fármacos , Fase II de la Desintoxicación Metabólica , Fase I de la Desintoxicación Metabólica , Acetaminofén/metabolismo , Analgésicos no Narcóticos/metabolismo , Antagonistas de Andrógenos/metabolismo , Transporte Biológico/efectos de los fármacos , Células Cultivadas , Cromatografía Líquida de Alta Presión , Medios de Cultivo Condicionados/química , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Sinergismo Farmacológico , Flutamida/análogos & derivados , Flutamida/metabolismo , Hepatocitos/metabolismo , Humanos , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Espectrometría de Masa por Ionización de Electrospray
6.
Chem Res Toxicol ; 19(10): 1341-50, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17040103

RESUMEN

Compound 1a (6-chloro-5-{3-[4-(1H-indazol-3-yl)-piperazin-1-yl]-propyl}-3,3-dimethyl-1,3-dihydro-indol-2-one) was mutagenic to Salmonella typhimurium TA98 in the presence of rat liver S9 subcellular fraction. The metabolism of 1a in rat liver S9 or microsomes demonstrated that it underwent a P450-mediated N-deindazolation (loss of indazole ring) as a predominant metabolic pathway. To investigate a possible link between metabolism and mutagenicity, a structural analogue 1b (6-chloro-5-{3-[4-(1H-indazol-3-yl)-piperidin-1-yl]-propyl}-3,3-dimethyl-1,3-dihydro-indol-2-one), the cleaved product 2a (6-chloro-3,3-dimethyl-5-(3-piperazin-1-yl-propyl)-1,3-dihydro-indol-2-one), and the core motif 3a (3-piperazinyl indazole) were evaluated in the Ames assay. It was found that 1b was not mutagenic to Salmonella typhimurium TA98 in the absence or presence of a metabolic activating system. In contrast to 1a, 1b did not undergo the metabolic cleavage (loss of indazole ring). Marginal mutagenicity of 2a to TA98 was observed with rat liver S9, whereas 3a was shown to be a promutagen. It was further demonstrated that 1a inactivated P450 3A, the principle enzyme catalyzing the N-deindazolation reaction, in an NADPH-, time-, and concentration-dependent manner. The kinetics of inactivation was characterized by a K(I) of 8.1 microM and k(inact) of 0.114 min(-1). The differences in mutagenicity between 1a and 1b suggest that a chemical bond extending from the 3-position of the indazole to a heteroatom (as part of another cyclic ring) is a prerequisite for the toxicity. The metabolic process leading to the elimination of the indazole from the rest of the molecule apparently plays a key role in causing mutagenicity. It is postulated that the N-deindazolation of 1a proceeds via an oxaziridine intermediate, the formation of which is indirectly inferred from the presence of benzoic acid in microsomal incubations. Benzoic acid is thought to be derived from the hydrolysis of 3-indazolone, an unstable product generated from the oxaziridine. Evidence suggests that the electrophilic oxaziridine intermediate may be responsible for the mutagenicity and inactivation of P450 3A.


Asunto(s)
Aziridinas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Indazoles/química , Mutagénesis/genética , Piperazinas/química , Animales , Aziridinas/química , Sistema Enzimático del Citocromo P-450/genética , Activación Enzimática/efectos de los fármacos , Indazoles/síntesis química , Indazoles/toxicidad , Cinética , Hígado/efectos de los fármacos , Hígado/metabolismo , Microsomas/efectos de los fármacos , Microsomas/metabolismo , Estructura Molecular , Oxidación-Reducción , Piperazinas/síntesis química , Piperazinas/toxicidad , Ratas , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/genética , Relación Estructura-Actividad , Testosterona/química , Testosterona/metabolismo
7.
Chem Res Toxicol ; 19(5): 701-9, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16696573

RESUMEN

The importance of uridine 5'-diphosphate-glucuronosyltranferases (UGT) 2B15 and other UGT enzymes (1A1, 1A6, and 1A9) in glucuronidating acetaminophen (APAP) is demonstrated. The kinetics and contributions of various UGTs in glucuronidating APAP are presented using clinically and toxicologically relevant concentrations of the substrate. UGT 1A9 and UGT 2B15 contribute significantly toward glucuronidating APAP when incubations were conducted in either phosphate or Tris-HCl buffers at 0.1 and 1.0 mM substrate concentrations. At 10 mM APAP, UGT 1A9 is a significant enzyme responsible for metabolizing APAP in either one of the buffers. UGT 1A1 is the next most important enzyme in glucuronidating APAP at this high substrate concentration. The contribution of UGT 1A6 at 10 mM APAP concentration became obscured by similar relative activities exhibited by UGTs 1A7, 1A8, and 2B7. These observations may reflect the differences in kinetic parameters for APAP glucuronidation by the individual UGTs. UGT 1A1 demonstrated Hill kinetics while UGT 1A9 displayed Michaelis-Menten kinetics. Substrate inhibition kinetics is observed with UGT 1A6, UGT 2B15, and human liver microsomes. The substrate inhibition is confirmed by employing stable isotope-labeled APAP as the substrate, while APAP glucuronide is used to test for inhibition of d4-APAP glucuronide. The in vitro hepatotoxicity caused by APAP in combination with phenobarbital or phenytoin is demonstrated in this study. The inhibition of APAP glucuronidation by phenobarbital leads to an increase in APAP-mediated toxicity in human hepatocytes. The toxicity to hepatocytes was further increased by coadministering APAP with phenytoin and phenobarbital. This synergistic increase in toxicity is postulated to be due to inhibition of UGTs (1A6, 1A9, and 2B15) responsible for detoxifying APAP through the glucuronidation pathway.


Asunto(s)
Acetaminofén/farmacocinética , Glucurónidos/metabolismo , Glucuronosiltransferasa/metabolismo , Analgésicos no Narcóticos/farmacocinética , Biotransformación , Células Cultivadas , Hepatocitos/metabolismo , Humanos , Microsomas Hepáticos/metabolismo
8.
Anal Chem ; 78(7): 2413-21, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16579628

RESUMEN

A rapid, sensitive, and specific assay for detection and quantitation of (p-chlorophenyl)aniline (CPA) in biological samples was developed. The assay was established based on rapid electrochemical oxidation of CPA to a dimerized product (1.0 V vs Pd) with the enhanced detection sensitivity of electrospray mass spectrometer (ES/MS). A "head-to-tail" dimer ([M + H]+ at m/z 217) was exhibited as the predominant species after electrochemical conversion of CPA. Optimal detection sensitivity and specificity for the dimer of CPA that was present in the biological matrix (e.g., rat urine) were achieved through on-line electrochemistry (EC) coupled with high-performance liquid chromatography tandem mass spectrometry. No matrix-associated ion suppression was observed. The limit of detection (S/N approximately 6) was 20 ng/mL, and the limit of quantitation was 50 ng/mL. The calibration curve was exhibited to be quadratic over the range of 50-2000 ng/mL with r2 > 0.99 in various biological matrixes. The assay was validated and used to study the biotransformation of p-chlorophenyl isocyanate (CPIC) to CPA in rats administered intraperitoneally with CPIC (50 mg/kg). The present LC/EC/MS/MS assay of CPA brings important technical advantages to assist in the risk assessment of new chemical entities, which have the potential to produce anilines via biotransformation.


Asunto(s)
Compuestos de Anilina/orina , Líquidos Corporales/efectos de los fármacos , Cromatografía Líquida de Alta Presión/métodos , Isocianatos/farmacocinética , Espectrometría de Masa por Ionización de Electrospray/métodos , Animales , Biotransformación , Líquidos Corporales/metabolismo , Calibración , Dimerización , Electroquímica , Inyecciones Intraperitoneales , Oxidación-Reducción , Ratas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
9.
Toxicol Sci ; 90(2): 451-9, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16410371

RESUMEN

Treatment with the antidepressant nefazodone has been associated with clinical idiosyncratic hepatotoxicty. Using membranes expressing human bile salt export pump (BSEP), human sandwich hepatocytes, and intact rats, we compared nefazodone and its marketed analogs, buspirone and trazodone. We found that nefazodone caused a strong inhibition of BSEP (IC(50) = 9 microM), inhibition of taurocholate efflux in human hepatocytes (IC(50) = 14 microM), and a transient increase in rat serum bile acids 1 h after oral drug administration. Buspirone or trazodone had no effect on biliary transport system. Nefazodone produced time- and concentration-dependent toxicity in human hepatocytes with IC(50) = 18 microM and 30 microM measured by inhibition of protein synthesis after 6 h and 24 h incubation, respectively. Toxicity was correlated with the amount of unmetabolized nefazodone. Partial recovery in toxicity by 24 h has been associated with metabolism of nefazodone to sulfate and glucuronide conjugates. The saturation of nefazodone metabolism resulted in sustained decrease in protein synthesis and cell death at 50 microM. The toxicity was not observed with buspirone or trazodone. Addition of 1-aminobenzotriazole (ABT), an inhibitor of CYP450, resulted in enhancement of nefazodone toxicity at 10 microM and was associated with accumulation of unmetabolized nefazodone. In human liver microsomes, ABT also prevented metabolism of nefazodone and formation of glutathione conjugates. We suggest that inhibition of bile acid transport by nefazodone is an indicator of potential hepatotoxicity. Our findings are consistent with the clinical experience and suggest that described methodology can be applied in the selection of nonhepatotoxic drug candidates.


Asunto(s)
Antidepresivos de Segunda Generación/toxicidad , Ácidos y Sales Biliares/metabolismo , Transporte Biológico/efectos de los fármacos , Triazoles/toxicidad , Animales , Ácidos y Sales Biliares/sangre , Canalículos Biliares/efectos de los fármacos , Canalículos Biliares/metabolismo , Buspirona/toxicidad , Células Cultivadas , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Piperazinas , Ratas , Ratas Sprague-Dawley , Trazodona/toxicidad
10.
Curr Drug Metab ; 6(3): 161-225, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15975040

RESUMEN

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.


Asunto(s)
Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Animales , Biotransformación/fisiología , Humanos , Microsomas Hepáticos/metabolismo , Compuestos Orgánicos/química , Compuestos Orgánicos/metabolismo , Relación Estructura-Actividad
11.
Chem Res Toxicol ; 16(3): 336-49, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12641434

RESUMEN

Capsaicin is a common dietary constituent and a popular homeopathic treatment for chronic pain. Exposure to capsaicin has been shown to cause various dose-dependent acute physiological responses including the sensation of burning and pain, respiratory depression, and death. In this study, the P450-dependent metabolism of capsaicin by recombinant P450 enzymes and hepatic and lung microsomes from various species, including humans, was determined. A combination of LC/MS, LC/MS/MS, and LC/NMR was used to identify several metabolites of capsaicin that were generated by aromatic (M5 and M7) and alkyl hydroxylation (M2 and M3), O-demethylation (M6), N- (M9) and alkyl dehydrogenation (M1 and M4), and an additional ring oxygenation of M9 (M8). Dehydrogenation of capsaicin was a novel metabolic pathway and produced unique macrocyclic, diene, and imide metabolites. Metabolism of capsaicin by microsomes was inhibited by the nonselective P450 inhibitor 1-aminobenzotriazole (1-ABT). Metabolism was catalyzed by CYP1A1, 1A2, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4. Addition of GSH (2 mM) to microsomal incubations stimulated the metabolism of capsaicin and trapped several reactive electrophilic intermediates as their GSH adducts. These results suggested that reactive intermediates, which inactivated certain P450 enzymes, were produced during catalytic turnover. Comparison of the rate and types of metabolites produced from capsaicin and its analogue, nonivamide, demonstrated similar pathways in the P450-dependent metabolism of these two capsaicinoids. However, production of the dehydrogenated (M4), macrocyclic (M1), and omega-1-hydroxylated (M3) metabolites was not observed for nonivamide. These differences may be reflective of the mechanism of formation of these metabolites of capsaicin. The role of metabolism in the cytotoxicity of capsaicin and nonivamide was also assessed in cultured lung and liver cells. Lung cells were markedly more sensitive to cytotoxicity by capsaicin and nonivamide. Cytotoxicity was enhanced 5 and 40% for both compounds by 1-ABT in BEAS-2B and HepG2, respectively. These data suggested that metabolism of capsaicinoids by P450 in cells represented a detoxification mechanism (in contrast to bioactivation).


Asunto(s)
Bronquios/metabolismo , Capsaicina/metabolismo , Capsaicina/toxicidad , Sistema Enzimático del Citocromo P-450/metabolismo , Hepatocitos/metabolismo , Animales , Bronquios/citología , Bronquios/efectos de los fármacos , Bronquios/enzimología , Capsaicina/química , Células Cultivadas , Inhibidores Enzimáticos del Citocromo P-450 , Inhibidores Enzimáticos/farmacología , Glutatión/química , Glutatión/farmacología , Cabras , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Humanos , Hidrogenación , Espectrometría de Masas/métodos , Ratones , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Resonancia Magnética Nuclear Biomolecular , Conejos , Ratas , Triazoles/farmacología
12.
Drug Metab Dispos ; 31(1): 122-32, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12485961

RESUMEN

(S)-5, 6-Difluoro-4-cyclopropylethynyl-4-trifluoromethyl-3, 4-dihydro- 2-(1H)-quinazolinone (DPC 963), a specific non-nucleoside inhibitor of human immunodeficiency virus-1 reverse transcriptase, is primarily metabolized in humans to the glucuronide conjugate of 8-OH DPC 963 (M8). Electrospray ionization-liquid chromatography/mass spectrometry analyses of urine from subjects dosed with DPC 963 also revealed the presence of other minor metabolites including glucuronide conjugate of 6-OH DPC 963 (M7). An oxidative defluorination pathway involving a putative p-benzoquinone imine capable of being reduced to the hydroquinone (M7) is postulated. The formation of the benzoquinone imine [detected as a glutathione (GSH) adduct, M5] was primarily carried out by CYP3A4, whereas M8 was formed mainly by the polymorphic CYP2B6. The kinetic studies with human liver microsomes showed that the apparent K(m) and V(max) values for the formation of M5 were 65.8 microM and 25.6 pmol/min/mg of protein, respectively. The formation of M8 showed K(m) and V(max) values of 15.1 microM and 22.9 pmol/min/mg of protein, respectively. The microsomal studies also revealed the occurrence of a possible oxirene intermediate that was trapped as GSH adducts M3 and M4. It was demonstrated, for the first time, that CYP3A4 was capable of directly oxidizing the triple bond of the cyclopropyl ethynyl group to an unstable oxirene. The apparent K(m) and V(max) values for the formation of an oxirene (detected as the GSH adduct M3) were 1.9 mM and 10.2 pmol/min/mg of protein, respectively. These results suggest that CYP2B6 has a higher affinity than CYP3A4 toward DPC 963. This consequently leads to greater levels of CYP2B6-catalyzed product, M8, than CYP3A4-mediated bioactivation of DPC 963 to benzoquinone imine or oxirene intermediates.


Asunto(s)
Microsomas Hepáticos/metabolismo , Quinolonas/metabolismo , Inhibidores de la Transcriptasa Inversa/metabolismo , Biotransformación/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Transcriptasa Inversa del VIH/antagonistas & inhibidores , Humanos , Cinética , Microsomas Hepáticos/enzimología , Quinolonas/química , Inhibidores de la Transcriptasa Inversa/química
13.
Chem Res Toxicol ; 15(9): 1190-207, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12230413

RESUMEN

The in vivo and in vitro disposition of benzylamine was investigated in rats. Benzylamine was metabolized to only a small extent by rat liver subcellular fractions. In contrast, it was extensively metabolized in vivo in rats. In vivo studies performed with stable isotope-labeled benzylamine enabled rapid mass spectrometric identification of metabolites present in rat bile and urine. The major metabolite of benzylamine was the hippuric acid formed by glycine conjugation of benzoic acid. LC/MS analysis of bile and urine obtained from rats dosed with 1:1 equimolar mixture of either d(0):d(7)- or d(0):d(2)-benzylamine showed the presence of several glutathione adducts in addition to the hippuric acid metabolite. The presence of various glutathione adducts indicated that benzylamine was metabolized to a number of reactive intermediates. Various metabolic pathways, including those independent of P450, were found to produce these intermediates. A previously undocumented pathway included the formation of a new carbon-nitrogen bond that led to a potentially reactive intermediate, Ar-CH(2)-NH(CO)-X, capable of interacting with various nucleophiles. The origin of this reactive intermediate is postulated to occur via the formation of either a formamide or carbamic acid metabolites. Metabolites which were produced by the reaction of this intermediate, Ar-CH(2)-NH(CO)-X with nucleophiles included S-[benzylcarbamoyl] glutathione, N-acetyl-S-[benzylcarbamoyl]cysteine, S-[benzylcarbamoyl] cysteinylglycine, S-[benzylcarbamoyl] cysteinylglutamate, N-[benzylcarbamoyl] glutamate, and an oxidized glutathione adduct. Bioactivation of amines via this pathway has not been previously described. The oxidative deamination of benzylamine yielding the benzaldehyde was demonstrated to be a precursor to the hippuric acid metabolite and S-benzyl-L-glutathione. The formation of the S-benzyl-L-glutathione conjugate showed that a net displacement of amine from benzylamine had taken place with a subsequent addition of glutathione at the benzylic position. In addition to these novel pathways, a number of other glutathione-derived adducts formed as a result of epoxide formation was characterized. It was demonstrated that benzylamine was converted by rat P450 2A1 and 2E1 to benzamide that was rapidly metabolized to an epoxide. Mechanisms are proposed for the formation of various GSH adducts of benzylamine.


Asunto(s)
Bencilaminas/farmacocinética , Glutamatos/biosíntesis , Glutatión/biosíntesis , Animales , Bilis/metabolismo , Biotransformación , Cromatografía Liquida , Sistema Enzimático del Citocromo P-450/clasificación , Sistema Enzimático del Citocromo P-450/metabolismo , Glutamatos/química , Glutamatos/orina , Glutatión/análogos & derivados , Glutatión/química , Glutatión/orina , Masculino , Espectrometría de Masas , Microsomas Hepáticos/metabolismo , Resonancia Magnética Nuclear Biomolecular/métodos , Oxidación-Reducción , Oximas/análisis , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/metabolismo
14.
Chem Res Toxicol ; 15(1): 48-62, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11800597

RESUMEN

The in vitro and in vivo disposition of DPC 423 was investigated in mice, rats, dogs and humans and the metabolites characterized by LC/MS, LC/NMR and high field-NMR. The rodents produced several metabolites that included an aldehyde (M1), a carboxylic acid (M2), a benzyl alcohol (M3), glutamate conjugates (M4 and M5), an acyl glucuronide (M6) and its isomers; a carbamyl glucuronide (M7); a phenol (M8) and its glucuronide conjugate (M9), two glutathione adducts (M10 and M11), a sulfamate conjugate (M12), isomers of an oxime metabolite (M13), and an amide (M14). Humans and dogs produced less complex metabolite profiles than rats. While unchanged DPC 423 was the major component in plasma and urine samples, differences in the metabolic disposition of this compound among species were noted. M1 is believed to be rapidly oxidized to the carboxylic acid (M2), which forms the potentially reactive acyl glucuronide (M6). The formation of novel glutamate conjugates (M4 and M5) and their role in depleting endogenous glutathione have been described previously. The carbamyl glucuronide M7, found as the major metabolite in rats and in other species, was considered nonreactive and was easily hydrolyzed to the parent compound in the presence of beta-glucuronidase. The identification of GSH adducts M10 and M11 led us to postulate the existence of at least two reactive intermediates responsible for their formation, an epoxide and possibly a nitrile oxide, respectively. Although the formation of GSH adducts such as M10 from epoxides has been described before, there are no reports to date describing the existence of a GSH adduct (M11) of an oxime. The formation of a sulfamate conjugate (M12) formed by direct coupling of sulfate to the nitrogen of benzylamine is described. A mechanism is proposed for the formation of the oxime (M13) that involves sequential oxidation of the benzylamine to the corresponding hydroxylamine and nitroso intermediate. The rearrangement of the nitroso intermediate is believed to produce the oxime (M13). In vitro studies suggested that both the oxime (M13) and the aldehyde (M1) were precursors to the carboxylic acid (M2). This is the first demonstration of carboxylic acid formation via an oxime intermediate produced from an amine. The stability of DPC423 in plasma obtained from several species was studied. Significant species differences in the plasma stability of DPC 423 were observed. The formation of the aldehyde metabolite (M1) was found to be catalyzed by a semicarbazide-sensitive monoamine oxidase (SSAO) found in plasma of rabbits, dogs, and rhesus monkeys. Rat, chimpanzee, and human plasma did not form M1.


Asunto(s)
Inhibidores del Factor Xa , Fibrinolíticos/farmacocinética , Pirazoles/farmacocinética , Sulfonas/farmacocinética , Adulto , Anciano , Animales , Cromatografía Líquida de Alta Presión , Perros , Fibrinolíticos/análisis , Humanos , Técnicas In Vitro , Espectroscopía de Resonancia Magnética , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos BALB C , Microsomas Hepáticos/metabolismo , Persona de Mediana Edad , Pirazoles/análisis , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Sulfonas/análisis
15.
Chem Res Toxicol ; 15(1): 63-75, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11800598

RESUMEN

The in vivo and in vitro disposition of DPC 423, a highly potent, selective, and orally bioavailable inhibitor of blood coagulation factor Xa, has recently been described. Several metabolites, some of which were considered potentially reactive, were identified in rats. A novel GSH adduct, the structure of which was not determined conclusively, was isolated from bile of rats dosed with DPC 423. Herein, we describe the complete structural elucidation of this unique GSH conjugate employing LC/MS and high-field NMR. Similar GSH adducts of DPC 602, [13CD2]DPC 602, and SX 737, all structural analogues of DPC 423, were isolated, characterized spectroscopically, and shown to have identical mass fragmentation pathways. The structures of these conjugates were initially suspected to be either an amide with N-S bond or a nitrogen-oxygen juxtaposed amide with a C-S bond. Studies conducted with [13CD2]DPC 602 indicated an aldoxime structure. The concluding evidence came from HMBC NMR spectrum of the conjugate, which showed strong correlation of the cysteine methylene protons with the imino carbon. Further spectroscopic studies with chemically prepared GSH adduct from benzaldehyde oxime confirmed this pattern of correlation. In vivo and in vitro studies with the synthetic oxime intermediate from DPC 423 showed an adduct identical to the one isolated from the bile of rats dosed with DPC 423. This supported the intermediacy of an aldoxime as a precursor to the GSH adducts. It is postulated that the benzylamine moiety of DPC 423 (and its analogues) is oxidized to a hydroxylamine, which is subsequently converted to a nitroso intermediate. Subsequent rearrangement of the nitroso leads to an aldoxime which in turn is metabolized by P450 to a reactive intermediate. The formation of oxime from DPC 423 (and its analogues) was found to be mediated by rat CYP 3A1/2, which were also responsible for converting the oxime to the GSH trappable reactive intermediate. It is postulated that the aldoxime produces a radical or a nitrile oxide intermediate that reacts with GSH and hence produces this unusual GSH adduct. On the basis of synthetic analogy, it is more likely that the nitrile oxide resulting from two-electron oxidation of the aldoxime is the reactive intermediate. Intramolecular kinetic isotope effects were studied with [13CD2]DPC 602 to assess the importance of the metabolic cleavage of the aminomethyl carbon-hydrogen bond in forming this GSH adduct. The lack of isotope effect in forming the aldoxime from [13CD2]DPC 602 suggests its formation does not occur through the imine intermediate. Instead the data supports the postulated mechanism of hydroxylamine and nitroso intermediates as precursors to the aldoxime. However, the formation of the GSH adduct from [13CD2]DPC 602 did show a significant intramolecular kinetic isotope effect (kH/kD = 2.3) since a carbon-deuterium bond had to be broken on the aldoxime prior to the formation of the adduct. A stable nitrile oxide derived from DPC 602 was postulated as the reactive intermediate responsible for forming this unique GSH adduct.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Inhibidores del Factor Xa , Fibrinolíticos/metabolismo , Glutatión/metabolismo , Oximas/metabolismo , Pirazoles/metabolismo , Sulfonas/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Fibrinolíticos/análisis , Masculino , Microsomas Hepáticos/metabolismo , Pirazoles/análisis , Ratas , Ratas Sprague-Dawley , Sulfonas/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...