Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Drug Metab Dispos ; 39(1): 30-8, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20926621

RESUMEN

Vorapaxar (SCH 530348), a potent oral thrombin protease-activated receptor 1 antagonist, is being developed as an antiplatelet agent for patients with established vascular disease. The objective of this study was to identify the human liver cytochrome P450 (P450) enzyme(s) responsible for the metabolism of SCH 530348. Human liver microsomes metabolized SCH 530348 to M19, an amine metabolite formed via carbamate cleavage, and M20 (monohydroxy-SCH 530348). Recombinant human CYP3A4 exhibited the most activity (11.5% profiled radioactivity) for the formation of M19, followed by markedly less substrate conversion with CYP1A1 and CYP2C19. Trace levels of M19, a major excreted human metabolite, were detected with CYP1A2, CYP3A5, and CYP4F3A. Formation of M19 by human liver microsomes was inhibited 89% by ketoconazole (IC(50), 0.73 µM), 34% by tranylcypromine, and 89% by anti-CYP3A4 monoclonal antibody. There was a significant correlation between the rate of M19 formation and midazolam 1'-hydroxylation (r = 0.75) or M19 formation and testosterone 6ß-hydroxylation (r = 0.92). The results of screening, inhibition, and correlation studies confirmed that CYP3A4 is the major P450 enzyme responsible for M19 formation from SCH 530348. In contrast, formation of M20, a major circulating human metabolite at steady state, was primarily catalyzed by CYP3A4 and CYP2J2. M20 is pharmacologically equipotent to SCH 530348, whereas M19 is an inactive metabolite. Formation of M20 by human liver microsomes was inhibited 89% by ketoconazole, 75% by astemizole (a CYP2J2 inhibitor), and 43% by CYP3A4 monoclonal antibody. These results suggest that CYP3A4 and CYP2J2 are both involved in the formation of M20 metabolite.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Lactonas/metabolismo , Piridinas/metabolismo , Receptor PAR-1/antagonistas & inhibidores , Receptores de Trombina/antagonistas & inhibidores , Astemizol/farmacología , Sistema Enzimático del Citocromo P-450/fisiología , Humanos , Hidroxilación , Cetoconazol/farmacología , Lactonas/sangre , Lactonas/farmacocinética , Lactonas/farmacología , Microsomas/metabolismo , Microsomas Hepáticos/metabolismo , Piridinas/sangre , Piridinas/farmacocinética , Piridinas/farmacología , Receptor PAR-1/metabolismo , Proteínas Recombinantes/metabolismo , Tranilcipromina/farmacología
2.
Drug Metab Dispos ; 39(3): 510-21, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21123164

RESUMEN

Boceprevir (SCH 503034), a protease inhibitor, is under clinical development for the treatment of human hepatitis C virus infections. In human liver microsomes, formation of oxidative metabolites after incubations with [(14)C]boceprevir was catalyzed by CYP3A4 and CYP3A5. In addition, the highest turnover was observed in recombinant CYP3A4 and CYP3A5. After a single radiolabeled dose to human, boceprevir was subjected to two distinct pathways, namely cytochrome P450-mediated oxidation and ketone reduction. Therefore, attempts were made to identify the enzymes responsible for the formation of carbonyl-reduced metabolites. Human liver S9 and cytosol converted ∼ 28 and ∼ 68% of boceprevir to M28, respectively, in the presence of an NADPH-generating system. Screening of boceprevir with recombinant human aldo-keto reductases (AKRs) revealed that AKR1C2 and AKR1C3 exhibited catalytic activity with respect to the formation of M+2 metabolites (M28 and M31). The formation of M28 was inhibited by 100 µM flufenamic acid (80.3%), 200 µM mefenamic acid (83.7%), and 100 µM phenolphthalein (86.1%), known inhibitors of AKRs, suggesting its formation through carbonyl reduction pathway. Formation of M28 was also inhibited by 100 µM diazepam (75.1%), 1 mM ibuprofen (70%), and 200 µM diflunisal (89.4%). These data demonstrated that CYP3A4 and CYP3A5 are primarily responsible for the formation of oxidative metabolites and the formation of M28 and M31, the keto-reduced metabolites, are most likely mediated by AKR1C2 and AKR1C3. Because the biotransformation and clearance of boceprevir involves two different enzymatic pathways, boceprevir is less likely to be a victim of significant drug-drug interaction with concomitant medication affecting either of these pathways.


Asunto(s)
Drogas en Investigación/metabolismo , Hepacivirus/enzimología , Hígado/enzimología , Prolina/análogos & derivados , Inhibidores de Serina Proteinasa/metabolismo , Proteínas no Estructurales Virales/antagonistas & inhibidores , 3-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas , Biotransformación/efectos de los fármacos , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A , Drogas en Investigación/química , Inhibidores Enzimáticos/farmacología , Humanos , Hidroxiprostaglandina Deshidrogenasas/antagonistas & inhibidores , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Hidroxiesteroide Deshidrogenasas/genética , Hidroxiesteroide Deshidrogenasas/metabolismo , Cinética , Estructura Molecular , Oligopéptidos/química , Oligopéptidos/metabolismo , Oxidación-Reducción , Prolina/química , Prolina/metabolismo , Proteínas Recombinantes/metabolismo , Inhibidores de Serina Proteinasa/química , Estereoisomerismo , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/enzimología , Fracciones Subcelulares/metabolismo , Urea/análogos & derivados , Urea/química , Urea/metabolismo
3.
Toxicol Appl Pharmacol ; 255(1): 48-56, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21640743

RESUMEN

An investigative renal toxicity study using metabolomics was conducted with a potent nicotinic acid receptor (NAR) agonist, SCH 900424. Liquid chromatography-mass spectrometry (LC-MS) and gas chromatography-mass spectrometry (GC-MS) techniques were used to identify small molecule biomarkers of acute kidney injury (AKI) that could aid in a better mechanistic understanding of SCH 900424-induced AKI in mice. The metabolomics study revealed 3-indoxyl sulfate (3IS) as a more sensitive marker of SCH 900424-induced renal toxicity than creatinine or urea. An LC-MS assay for quantitative determination of 3IS in mouse matrices was also developed. Following treatment with SCH 900424, 3IS levels were markedly increased in murine plasma and brain, thereby potentially contributing to renal- and central nervous system (CNS)-related rapid onset of toxicities. Furthermore, significant decrease in urinary excretion of 3IS in those animals due to compromised renal function may be associated with the elevation of 3IS in plasma and brain. These data suggest that 3IS has a potential to be a marker of renal and CNS toxicities during chemically-induced AKI in mice. In addition, based on the metabolomic analysis other statistically significant plasma markers including p-cresol-sulfate and tryptophan catabolites (kynurenate, kynurenine, 3-indole-lactate) might be of toxicological importance but have not been studied in detail. This comprehensive approach that includes untargeted metabolomic and targeted bioanalytical sample analyses could be used to investigate toxicity of other compounds that pose preclinical or clinical development challenges in a pharmaceutical discovery and development.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Encéfalo/metabolismo , Indicán/análisis , Metabolómica , Agonistas Nicotínicos/toxicidad , Lesión Renal Aguda/metabolismo , Animales , Biomarcadores , Indicán/sangre , Riñón/efectos de los fármacos , Masculino , Ratones , Proteína 1 de Transporte de Anión Orgánico/fisiología , Transportadores de Anión Orgánico Sodio-Independiente/fisiología
4.
Anal Chem ; 82(24): 10251-7, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21105697

RESUMEN

Recent FDA and ICH guidances on safety testing of drug metabolites have challenged the way we traditionally think about quantitative bioanalytical methods. Such assays, in general, require a reference standard for each analyte to construct calibration curves and prepare quality control samples. However, early in the drug development process, metabolite standards may not be readily available, and if they are inherently unstable, they are difficult to synthesize or purify. In this paper, we describe a novel in-line method for producing and then quantifying a very unstable metabolite which is based upon the in situ postcolumn coulometric oxidation of the parent drug. Lacking any metabolite standards, the feasibility of simultaneously quantifying a development drug (compound A) and its unstable hydroxylated metabolites (metabolite B) was investigated. Reference standards for these ostensibly major human metabolites could not be reliably obtained due to rapid degradation upon purification and/or subsequent storage. Following high-performance liquid chromatography (HPLC) separation, parent drug and its [(13)C(3)-(15)N] isotopically labeled internal standard were quantitatively converted to equal amounts of a diastereomeric pair of hydroxylated metabolites using a postcolumn coulometric electrochemical cell before reaching the mass spectrometer. The concentration of the injected parent (which is equal to the total concentration of the in-line generated metabolites since the conversion to metabolite is quantitative) and the tandem mass spectrometry (MS/MS) signals of the electrochemically generated metabolites were used to construct a calibration curve for quantifying both the parent drug and its hydroxylated metabolites. Plasma extracts from humans dosed with compound A contained chromatographically distinct liquid chromatography-mass spectrometry (LC-MS) signals (m/z 538) for in vivo formed hydroxylated metabolites and the electrochemically oxidized parent drug which had been converted in-line into its chemically identical twin. Both peaks in this study sample could be quantified using a single calibration curve obtained under the same coulometric conditions using known amounts of the parent drug. Although no attempt was made to fully validate a bioanalytical method, the practicality of this in situ quantification approach was further confirmed by the preliminary bioanalytical analysis of a selection of plasma samples collected following oral administration (50 mg) of compound A in a clinical study.


Asunto(s)
Cromatografía Liquida , Preparaciones Farmacéuticas/análisis , Preparaciones Farmacéuticas/metabolismo , Espectrometría de Masas en Tándem , Isótopos de Carbono , Humanos , Hidroxilación , Isótopos de Nitrógeno , Preparaciones Farmacéuticas/sangre
5.
Drug Metab Dispos ; 38(11): 2067-74, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20671096

RESUMEN

The study of human metabolism of endo-8[bis(2-chlorophenyl)methyl]-3-(2-pyrimidinyl)-8-azabicyclo[3.2.1]octan-3-ol (SCH 486757) after a 200-mg oral dose of the drug to healthy volunteers in the first-in-human study is presented. The structural elucidation of two unique metabolites, which were detected in the process of metabolite characterization in human plasma and urine by liquid chromatography-mass spectrometry (LC-MS), is described. These metabolites (M27 and M34) were initially detected in human plasma at high levels (>35% of the LC-MS response of the parent drug). Additional LC-MS experiments (hydrogen/deuterium exchange and accurate mass measurement) were used to determine structures of metabolites. It was found that both metabolites were formed through a loss of the C-C bridge from the tropane moiety with the conversion into a substituted pyridinium compound. This metabolic process has not been reported previously. Because of the apparent high abundance of metabolites based on the LC-MS response, actual circulating amounts of these metabolites relative to the parent drug were determined semiquantitatively to evaluate their coverage in preclinical species. With the use of reference standards, it was shown that the LC-MS response of M27 and M34 in human plasma was much higher than that of the parent compound. Actual amounts of M27 and M34 metabolites were less than 5% of the level of the parent drug; therefore, additional assessment was not required.


Asunto(s)
Antitusígenos/metabolismo , Compuestos de Azabiciclo/metabolismo , Compuestos de Piridinio/metabolismo , Pirimidinas/metabolismo , Receptores Opioides/agonistas , Animales , Antitusígenos/sangre , Antitusígenos/farmacocinética , Antitusígenos/farmacología , Antitusígenos/orina , Compuestos de Azabiciclo/sangre , Compuestos de Azabiciclo/farmacocinética , Compuestos de Azabiciclo/farmacología , Compuestos de Azabiciclo/orina , Biotransformación , Cromatografía Líquida de Alta Presión , Humanos , Masculino , Conformación Molecular , Compuestos de Piridinio/sangre , Compuestos de Piridinio/química , Compuestos de Piridinio/orina , Pirimidinas/sangre , Pirimidinas/química , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Pirimidinas/orina , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Receptor de Nociceptina
6.
J Am Soc Mass Spectrom ; 18(10): 1891-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17766144

RESUMEN

The widely different LC-MS response observed for many structurally different compounds limits the use of LC-MS in full scan detection mode for quantitative determination of drugs and metabolites without using reference standard. The recently introduced nanospray ionization (NSI) technique shows comparable MS response for some compounds under non-LC-MS conditions. However, in the presence of numerous endogenous compounds commonly associated with biological samples such as urine, plasma, and bile, LC-MS is required to separate, detect, identify, and measure individual analytes. An LC-NSI-MS system was devised and the MS response obtained in this system for a variety of pharmaceutical drugs and their metabolites. The set-up involves two high-performance liquid chromatography (HPLC) systems, a chip-based NSI source and a quadrupole-time-of-flight (Q-TOF) mass spectrometer. Herein this is referred to as the response normalized-liquid chromatography NSI-MS (RNLC-NSI-MS) system. One HPLC unit performs the analytical separation, while the other unit adds solvent post-column with an exact reverse of the mobile phase composition such that the final composition entering the NSI source is isocratic throughout the entire HPLC run. The data obtained from four different structural classes of compounds [vicriviroc (VCV), desloratadine (DL), tolbutamide, and cocaine] and their metabolites indicate that by maintaining the solvent composition unchanged across the HPLC run, the influence of the solvent environment on the ionization efficiency is minimized. In comparison to responses obtained from radiochromatograms, responses from conventional LC-ESI-MS overestimated the VCV and DL responses, respectively, by 6- and 20-fold. Although VCV and DL responses obtained using LC-NSI-MS are within 2- to 6-fold from the respective radiochromatographic responses, the response normalization modification results in nearly uniform LC-NSI-MS response for all compounds evaluated.


Asunto(s)
Algoritmos , Cromatografía Líquida de Alta Presión/métodos , Microquímica/métodos , Nanotecnología/métodos , Espectrometría de Masa por Ionización de Electrospray/métodos , Urinálisis/métodos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
7.
Curr Drug Metab ; 7(5): 503-23, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16787159

RESUMEN

Metabolism studies play a pivotal role in drug discovery and development. Characterization of metabolic "hot-spots" as well as reactive and pharmacologically active metabolites is critical to designing new drug candidates with improved metabolic stability, toxicological profile and efficacy. Metabolite identification in the preclinical species used for safety evaluation is required in order to determine whether human metabolites have been adequately tested during non-clinical safety assessment. From an instrumental standpoint, high performance liquid chromatography (HPLC) coupled with mass spectrometry (MS) dominates all analytical tools used for metabolite identification. The general strategies employed for metabolite identification in both drug discovery and drug development settings together with sample preparation techniques are reviewed herein. These include a discussion of the various ionization methods, mass analyzers, and tandem mass spectrometry (MS/MS) techniques that are used for structural characterization in a modern drug metabolism laboratory. Mass spectrometry-based techniques, such as stable isotope labeling, on-line H/D exchange, accurate mass measurement to enhance metabolite identification and recent improvements in data acquisition and processing for accelerating metabolite identification are also described. Rounding out this review, we offer additional thoughts about the potential of alternative and less frequently used techniques such as LC-NMR/MS, CRIMS and ICPMS.


Asunto(s)
Biotransformación , Preparaciones Farmacéuticas/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Animales , Cromatografía Líquida de Alta Presión/métodos , Medición de Intercambio de Deuterio , Análisis de Fourier , Humanos , Almacenamiento y Recuperación de la Información , Marcaje Isotópico , Espectroscopía de Resonancia Magnética/métodos , Espectrometría de Masas/métodos , Espectrometría de Masas/tendencias , Microquímica/métodos , Estructura Molecular , Peso Molecular , Preparaciones Farmacéuticas/química , Manejo de Especímenes/métodos , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Tecnología Farmacéutica/métodos , Tecnología Farmacéutica/tendencias
8.
J Mass Spectrom ; 41(11): 1430-41, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17051523

RESUMEN

Structural characterization of unstable metabolites and other drug-derived entities poses a serious challenge to the analytical chemist using instrumentation such as LC-MS and LC-MS/MS, and may lead to inaccurate identification of metabolite structures. The task of structural elucidation becomes even more difficult when an analyte is unstable in the ion source of the mass spectrometer. However, a judicious selection of the experimental conditions and the advanced features of new generation mass spectrometers can often overcome these difficulties. We describe here the identification of three drug-derived peaks (A, B and C) that were detected from a Schering-Plough developmental compound (Lonafarnib) following incubation with cDNA-expressed human CYP3A4. Definitive characterization was achieved using (1) accurate mass measurement, (2) stable isotope incorporation, (3) reduced ion source temperature, (4) alkali ion attachment and (5) MS/MS fragmentation studies. The protonated ions of compounds A and B fragmented almost completely in the source, yielding ions of the same mass-to-charge ratio (m/z) as that of protonated C (CH+). Fortunately, the presence of Na+ and K+ adducts of A and B provided information crucial to distinguishing AH+ and BH+ from their fragment ions. Metabolite A was shown to be an unstable hydroxylated metabolite of Lonafarnib. The metabolite C was shown to be a dehydrogenated metabolite of Lonafarnib (Lonafarnib-2H), unstable in the presence of protic solvents. Finally, B was artifactually formed most likely from C by the solvolytic addition of methanol during sample preparation. MS/MS fragmentation experiments assisted in identifying the site of metabolism in A and chemical modification in B. A and C readily interconvert through hydration/dehydration, and B and C through addition/elimination of methanol present in the sample-processing solvents. Finally, NMR experiments were performed to confirm the structures of A and C.


Asunto(s)
Cromatografía Liquida/métodos , Espectrometría de Masas/métodos , Piperidinas/metabolismo , Piridinas/metabolismo , Temperatura , Isótopos de Carbono , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/metabolismo , Estabilidad de Medicamentos , Humanos , Iones/química , Marcaje Isotópico , Espectroscopía de Resonancia Magnética , Estructura Molecular
9.
Clin Pharmacokinet ; 44(5): 467-94, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15871634

RESUMEN

Ezetimibe is the first lipid-lowering drug that inhibits intestinal uptake of dietary and biliary cholesterol without affecting the absorption of fat-soluble nutrients. Following oral administration, ezetimibe is rapidly absorbed and extensively metabolised (>80%) to the pharmacologically active ezetimibe-glucuronide. Total ezetimibe (sum of 'parent' ezetimibe plus ezetimibe-glucuronide) concentrations reach a maximum 1-2 hours post-administration, followed by enterohepatic recycling and slow elimination. The estimated terminal half-life of ezetimibe and ezetimibe-glucuronide is approximately 22 hours. Consistent with the elimination half-life of ezetimibe, an approximate 2-fold accumulation is observed upon repeated once-daily administration. The recommended dose of ezetimibe 10 mg/day can be administered in the morning or evening without regard to food. There are no clinically significant effects of age, sex or race on ezetimibe pharmacokinetics and no dosage adjustment is necessary in patients with mild hepatic impairment or mild-to-severe renal insufficiency. The major metabolic pathway for ezetimibe consists of glucuronidation of the 4-hydroxyphenyl group by uridine 5'-diphosphate-glucuronosyltransferase isoenzymes to form ezetimibe-glucuronide in the intestine and liver. Approximately 78% of the dose is excreted in the faeces predominantly as ezetimibe, with the balance found in the urine mainly as ezetimibe-glucuronide. Overall, ezetimibe has a favourable drug-drug interaction profile, as evidenced by the lack of clinically relevant interactions between ezetimibe and a variety of drugs commonly used in patients with hypercholesterolaemia. Ezetimibe does not have significant effects on plasma levels of HMG-CoA reductase inhibitors commonly known as statins (atorvastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin, simvastatin), fibric acid derivatives (gemfibrozil, fenofibrate), digoxin, glipizide, warfarin and triphasic oral contraceptives (ethinylestradiol and levonorgestrel). Concomitant administration of food, antacids, cimetidine or statins had no significant effect on ezetimibe bioavailability. Although coadministration with gemfibrozil and fenofibrate increased the bioavailability of ezetimibe, the clinical significance is thought to be minor considering the relatively flat dose-response curve of ezetimibe and the lack of dose-related increase in adverse events. In contrast, coadministration with the bile acid binding agent colestyramine significantly decreased ezetimibe oral bioavailability (based on area under the plasma concentration-time curve of total ezetimibe). Hence, ezetimibe and colestyramine should be administered several hours apart to avoid attenuating the efficacy of ezetimibe. Finally, higher ezetimibe exposures were observed in patients receiving concomitant ciclosporin, and ezetimibe caused a small but statistically significant effect on plasma levels of ciclosporin. Because treatment experience in patients receiving ciclosporin is limited, physicians are advised to exercise caution when initiating ezetimibe in the setting of ciclosporin coadministration, and to carefully monitor ciclosporin levels.


Asunto(s)
Anticolesterolemiantes/farmacocinética , Azetidinas/farmacocinética , Animales , Azetidinas/farmacología , Disponibilidad Biológica , Resina de Colestiramina/farmacología , Anticonceptivos Orales/farmacología , Interacciones Farmacológicas , Ezetimiba , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Warfarina/farmacología
10.
Artículo en Inglés | MEDLINE | ID: mdl-12016032

RESUMEN

A rapid HPLC method was developed for quantification of unbound evernimicin in human plasma. Protein-free samples prepared by ultrafiltration were injected directly onto a polymeric reversed-phase column and the eluent monitored at 302 nm. Evernimicin that eluted within 3.5 min was well resolved from endogenous components. Linearity was established between peak height and evernimicin concentration from 25 to 2500 ng/ml. Assay precision (C.V.) was within 5% while bias was no greater than 3%. This method has been used for the ex vivo assessment of evernimicin protein binding in human plasma from safety and tolerance as well as liver dysfunction and renal insufficiency studies.


Asunto(s)
Aminoglicósidos , Antibacterianos/sangre , Cromatografía Líquida de Alta Presión/métodos , Humanos , Reproducibilidad de los Resultados , Espectrofotometría Ultravioleta , Ultrafiltración
11.
Adv Drug Deliv Rev ; 63(10-11): 923-42, 2011 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-21689702

RESUMEN

Intestinal lymphatic transport has been shown to be an absorptive pathway following oral administration of lipids and an increasing number of lipophilic drugs, which once absorbed, diffuse across the intestinal enterocyte and while in transit associate with secretable enterocyte lipoproteins. The chylomicron-associated drug is then secreted from the enterocyte into the lymphatic circulation, rather than the portal circulation, thus avoiding the metabolically-active liver, but still ultimately returning to the systemic circulation. Because of this parallel and potentially alternative absorptive pathway, first-pass metabolism can be reduced while increasing lymphatic drug exposure, which opens the potential for novel therapeutic modalities and allows the implementation of lipid-based drug delivery systems. This review discusses the physiological features of the lymphatics, enterocyte uptake and metabolism, links between drug transport and lipid digestion/re-acylation, experimental model (in vivo, in vitro, and in silico) of lymphatic transport, and the design of lipid- or prodrug-based drug delivery systems for enhancing lymphatic drug transport.


Asunto(s)
Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Sistema Linfático/metabolismo , Administración Oral , Animales , Transporte Biológico , Quilomicrones/metabolismo , Humanos , Lípidos/administración & dosificación , Lípidos/química , Lípidos/farmacocinética , Profármacos
12.
Drug Metab Lett ; 3(3): 162-70, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19702548

RESUMEN

The present study demonstrated that in addition to CYP3A4 and CYP2D6, the metabolism of loratadine is also catalyzed by CYP1A1, CYP2C19, and to a lesser extent by CYP1A2, CYP2B6, CYP2C8, CYP2C9 and CYP3A5. The biotransformation of loratadine was associated with the formation of desloratadine (DL) and further hydroxylation of both DL and the parent drug (loratadine). Based on the inhibition and correlation studies contribution of CYP2C19 in the formation of the major circulating metabolite DL seems to be minor. Reported clinical results suggest that the steady state mean (%CV) plasma Cmax and AUC(24hr) of loratadine were 4.73 ng/ml (119%) and 24.1 ng.hr/ml (157%), respectively, after dosing with 10 mg loratadine tablets for 10 days. High inter-subject variability in loratadine steady-state data is probably due to the phenotypical characteristics of CYP2D6, CYP2C19, and CYP3A4. The relative abundance of CYP3A4 in the human liver exceeds that of CYP2C19 and CYP2D6 and therefore the contribution of CYP3A4 in the metabolism of loratadine should be major (approximately 70%).


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Antagonistas de los Receptores Histamínicos H1 no Sedantes/metabolismo , Hígado/enzimología , Loratadina/metabolismo , Administración Oral , Biotransformación , Cromatografía Líquida de Alta Presión , Inhibidores Enzimáticos del Citocromo P-450 , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Antagonistas de los Receptores Histamínicos H1 no Sedantes/administración & dosificación , Antagonistas de los Receptores Histamínicos H1 no Sedantes/farmacocinética , Humanos , Hidroxilación , Isoenzimas/metabolismo , Cetoconazol/farmacología , Hígado/efectos de los fármacos , Loratadina/administración & dosificación , Loratadina/análogos & derivados , Loratadina/farmacocinética , Microsomas Hepáticos/enzimología , Proteínas Recombinantes/metabolismo , Comprimidos , Espectrometría de Masas en Tándem
13.
Drug Metab Dispos ; 35(12): 2186-95, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17827338

RESUMEN

Vicriviroc (SCH 417690), a CCR5 receptor antagonist, is currently under investigation for the treatment of human immunodeficiency virus infection. The objective of this study was to identify human liver cytochrome P450 enzyme(s) responsible for the metabolism of vicriviroc. Human liver microsomes metabolized vicriviroc via N-oxidation (M2/M3), O-demethylation (M15), N,N-dealkylation (M16), N-dealkylation (M41), and oxidation to a carboxylic acid metabolite (M35b/M37a). Recombinant human CYP3A4 catalyzed the formation of all these metabolites, whereas CYP3A5 catalyzed the formation of M2/M3 and M41. CYP2C9 only catalyzed the formation of M15. There was a high correlation between the rates of formation of M2/M3, M15, and M41, which was determined using 10 human liver microsomal samples and testosterone 6beta-hydroxylation catalyzed by CYP3A4/5 (r > or = 0.91). Ketoconazole and azamulin (inhibitors of CYP3A4) were potent inhibitors of the formation of M2/M3, M15, M41, and M35b/M37a from human liver microsomes. A CYP3A4/5-specific monoclonal antibody (1 microg/microg of protein) inhibited the formation of all metabolites from human liver microsomes by 86 to 100%. The results of this study suggest that formation of the major vicriviroc metabolites in human liver microsomes is primarily mediated via CYP3A4. CYP2C9 and CYP3A5 most likely play a minor role in the biotransformation of this compound. These enzymology data will provide guidance to design clinical studies to address any potential drug-drug interactions.


Asunto(s)
Antagonistas de los Receptores CCR5 , Ácidos Carboxílicos/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Factores Inmunológicos/metabolismo , Hígado/enzimología , Piperazinas/metabolismo , Pirimidinas/metabolismo , Anticuerpos Monoclonales , Hidrocarburo de Aril Hidroxilasas/metabolismo , Biotransformación , Hidrocarburos Aromáticos con Puentes/farmacología , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP3A , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/inmunología , Remoción de Radical Alquila , Inhibidores Enzimáticos/farmacología , Humanos , Factores Inmunológicos/farmacología , Técnicas In Vitro , Isoenzimas/metabolismo , Cetoconazol/farmacología , Cinética , Hígado/efectos de los fármacos , Oxidación-Reducción , Piperazinas/farmacología , Pirimidinas/farmacología , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Testosterona/metabolismo , Triazoles/farmacología
14.
Anal Chem ; 77(11): 3676-82, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15924404

RESUMEN

N-Oxides are known to undergo deoxygenation during atmospheric pressure chemical ionization (Ramanathan, R.; Su, A.-D.; Alvarez, N.; Blumenkrantz, N.; Chowdhury, S. K.; Alton, K.; Patrick, J. Anal. Chem. 2000, 72, 1352-1359) resulting from thermal energy activation at the vaporizer of the APCI source. In addition to deoxygenation, tert-N-oxides containing an alkyl or benzyl group on the N-oxide nitrogen also undergo an N-R to O-R rearrangement (Meisenheimer arrangement, where R = alkyl or benzyl), followed by elimination of an aldehyde (or a ketone) through an internal hydrogen transfer. This has been observed under both atmospheric pressure chemical ionization and atmospheric pressure photoionization conditions. These fragment ions were not observed in the product ion spectra from the protonated molecules of the corresponding N-oxides. The elimination of an aldehyde or a ketone, thus, results from thermal energy activation at the vaporizer and is not induced by collisional activation. These fragmentations not only distinguish N-oxides from isomeric hydroxylated metabolites but also provide a potential way to determine the position of N-oxidation when a metabolite (or molecule) contains multiple N-oxidation sites that are in different chemical environments.


Asunto(s)
Óxidos N-Cíclicos/química , Espectrometría de Masas/métodos , Temperatura , Presión Atmosférica , Derivados de Atropina/química , Clozapina/análogos & derivados , Clozapina/química , Etilaminas/química , Hidroxilación , Estructura Molecular , Oxidación-Reducción , Fotoquímica , Piridinas/química , Sensibilidad y Especificidad
15.
Biopharm Drug Dispos ; 25(6): 243-52, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15334623

RESUMEN

Desloratadine is a non-sedating antihistamine recently approved for the treatment of seasonal allergic rhinitis. The major metabolite of desloratadine in human plasma and urine is the glucuronide conjugate of 3-hydroxydesloratadine. 3-Hydroxydesloratadine-glucuronide is also the major in vitro metabolite of 3-hydroxydesloratadine formed by incubation of 3-hydroxydesloratadine with human liver microsomes supplemented with uridine 5'-diphosphate-glucuronic acid (UDPGA). The metabolite structure was confirmed by LC-MS and LC-MS/MS. Out of ten recombinant human UDP-glucuronosyltransferases (UGTs), UGT1A1, UGT1A3, UGT1A8 and UGT2B15 exhibited catalytic activity with respect to the formation of 3-hydroxydesloratadine-glucuronide. Inhibition studies with known inhibitors of UGT (diclofenac, flunitrazepam and bilirubin) confirmed the involvement of UGT1A1, UGT1A3 and UGT2B15 in the formation of 3-hydroxydesloratadine-glucuronide. The results from this study demonstrated that the in vitro formation of 3-hydroxydesloratadine-glucuronide from 3-hydroxydesloratadine was mediated via UGT1A1, UGT1A3 and UGT2B15 in human liver.


Asunto(s)
Glucurónidos/metabolismo , Glucuronosiltransferasa/metabolismo , Antagonistas de los Receptores Histamínicos H1 no Sedantes/metabolismo , Loratadina/análogos & derivados , Loratadina/metabolismo , Bilirrubina/farmacología , Diclofenaco/farmacología , Inhibidores Enzimáticos/farmacología , Flunitrazepam/farmacología , Glucuronosiltransferasa/administración & dosificación , Glucuronosiltransferasa/antagonistas & inhibidores , Humanos , Técnicas In Vitro , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Factores de Tiempo
16.
Drug Metab Dispos ; 30(4): 430-7, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11901097

RESUMEN

Ezetimibe [SCH 58235; 1-(4-fluorophenyl)-3(R)-[3-(4-fluorophenyl)-3(S)-hydroxypropyl]-4(S)-(4-hydroxyphenyl)-2-azetidinone], a selective cholesterol absorption inhibitor, is being developed for the treatment of primary hypercholesterolemia. The absorption, metabolism, and excretion of ezetimibe were characterized in eight healthy male volunteers in this single-center, single-dose, open-label study. Subjects received a single oral 20-mg dose of [14C]ezetimibe (approximately 100 microCi) with 200 ml of noncarbonated water after a 10-h fast. Concentrations of radioactivity and/or ezetimibe (conjugated and unconjugated) were determined in plasma, urine, and fecal samples. Ezetimibe was rapidly absorbed and extensively conjugated following oral administration. The main circulating metabolite in plasma was SCH 60663 [1-O-[4-[trans-(2S,3R)-1-(4-fluorophenyl)-4-oxo-3-[3(S)-hydroxy-3-(4-fluorophenyl)propyl]-2-azetidinyl]phenyl]-beta-D-glucuronic acid], the glucuronide conjugate of ezetimibe. Plasma concentration-time profiles of unconjugated and conjugated drug exhibited multiple peaks, indicating enterohepatic recycling. Approximately 78 and 11% of the administered [14C]ezetimibe dose were excreted in feces and urine, respectively, by 240 h after drug administration. Total recovery of radioactivity averaged 89% of the administered dose. The main excreted metabolite was the glucuronide conjugate of ezetimibe. The primary metabolite in urine (0- to72-h composite) was also the glucuronide conjugate (about 9% of the administered dose). Significant amounts (69% of the dose) of ezetimibe were present in the feces, presumably as a result of SCH 60663 hydrolysis and/or unabsorbed drug. No adverse events were reported in this study. A single 20-mg capsule of [(14)C]ezetimibe was safe and well tolerated after oral administration. The pharmacokinetics of ezetimibe are consistent with extensive glucuronidation and enterohepatic recirculation. The primary metabolic pathway for ezetimibe is by glucuronidation of the 4-hydroxyphenyl group.


Asunto(s)
Anticolesterolemiantes/metabolismo , Anticolesterolemiantes/farmacocinética , Azetidinas/metabolismo , Azetidinas/farmacocinética , Adulto , Anticolesterolemiantes/análisis , Área Bajo la Curva , Azetidinas/análisis , Azetidinas/sangre , Azetidinas/orina , Biotransformación , Cromatografía Liquida , Circulación Enterohepática , Ezetimiba , Heces/química , Humanos , Masculino , Espectrometría de Masas , Orina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA