Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 317(2): 579-89, 2006 May.
Article in English | MEDLINE | ID: mdl-16421286

ABSTRACT

The multidrug resistance protein Mrp2 is an ATP-binding cassette (ABC) transporter mainly expressed in liver, kidney, and intestine. One of the physiological roles of Mrp2 is to transport bilirubin glucuronides from the liver into the bile. Current in vivo models to study Mrp2 are the transporter-deficient and Eisai hyperbilirubinemic rat strains. Previous reports showed hyperbilirubinemia and induction of Mrp3 in the hepatocyte sinusoidal membrane in the mutant rats. In addition, differences in liver cytochrome P450 and UGT1a levels between wild-type and mutant rats were detected. To study whether these compensatory mechanisms were specific to rats, we characterized Mrp2(-/-) mice. Functional absence of Mrp2 in the knockout mice was demonstrated by showing increased levels of bilirubin and bilirubin glucuronides in serum and urine, a reduction in biliary excretion of bilirubin glucuronides and total glutathione, and a reduction in the biliary excretion of the Mrp2 substrate dibromosulfophthalein. To identify possible compensatory mechanisms in Mrp2(-/-) mice, the expression levels of 98 phase I, phase II, and transporter genes were compared in liver, kidney, and intestine of male and female Mrp2(-/-) and control mice. Unlike in Mrp2 mutant rats, no induction of Mrp3 in Mrp2(-/-) mice was detected. However, Mrp4 mRNA and protein in liver and kidney were increased approximately 6- and 2-fold, respectively. Phenotypic analysis of major cytochrome P450-mediated activities in liver microsomes did not show differences between wild-type and Mrp2(-/-) mice. In conclusion, Mrp2(-/-) mice are a new valuable tool to study the role of Mrp2 in drug disposition.


Subject(s)
Bilirubin/analogs & derivatives , Gene Expression , Membrane Transport Proteins/genetics , Multidrug Resistance-Associated Proteins/genetics , Animals , Bile/metabolism , Bilirubin/blood , Bilirubin/urine , Cytochrome P-450 Enzyme System/metabolism , Female , Glutathione/metabolism , Intestinal Mucosa/metabolism , Kidney/metabolism , Liver/metabolism , Male , Metabolic Clearance Rate , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Sulfobromophthalein/pharmacokinetics
2.
Drug Metab Dispos ; 34(1): 145-51, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16251255

ABSTRACT

Although zomepirac (ZP) and tolmetin (TM) induce anaphylactic reactions and form reactive acyl glucuronides, a direct link between the two events remains obscure. We report herein that, in addition to acyl glucuronidation, both drugs are subject to oxidative bioactivation. Following incubations of ZP with human liver microsomes fortified with NADPH and glutathione (GSH), a metabolite with an MH+ ion at m/z 597 was detected by LC/MS/MS. On the basis of collision-induced dissociation and NMR evidence, the structure of this metabolite was determined to be 5-[4'-chlorobenzoyl]-1,4-dimethyl-3-glutathionylpyrrole-2-acetic acid (ZP-SG), suggesting that the pyrrole moiety of ZP had undergone oxidation to an epoxide intermediate, followed by addition of GSH and loss of the elements of H2O to yield the observed conjugate. The oxidative bioactivation of ZP most likely is catalyzed by cytochrome P450 (P450) 3A4, since the formation of ZP-SG was reduced to approximately 10% of control values following pretreatment of human liver microsomes with ketoconazole or with an inhibitory anti-P450 3A4 IgG. A similar GSH adduct, namely 5-[4'-methylbenzoyl]-1-methyl-3-glutathionylpyrrole-2-acetic acid (TM-SG), was identified when TM was incubated with human liver microsomal preparations. The relevance of these in vitro findings to the in vivo situation was established through the detection of the same thiol adducts in rats treated with ZP and TM, respectively. Taken together, these data suggest that, in addition to the formation of acyl glucuronides, oxidative metabolism of ZP and TM affords reactive species that may haptenize proteins and thereby contribute to the drug-mediated anaphylactic reactions.


Subject(s)
Glutathione/metabolism , Microsomes, Liver/metabolism , Tolmetin/analogs & derivatives , Tolmetin/metabolism , Animals , Chromatography, Liquid/methods , Female , Glutathione/chemistry , Glutathione/pharmacology , Hepatocytes/chemistry , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Male , Microsomes, Liver/chemistry , Microsomes, Liver/drug effects , NADP/metabolism , NADP/pharmacology , Oxidation-Reduction/drug effects , Rats , Spectrometry, Mass, Electrospray Ionization/methods , Tritium , Troleandomycin/metabolism , Troleandomycin/pharmacology
3.
Drug Metab Dispos ; 32(9): 909-14, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15319330

ABSTRACT

Genetically obese Zucker rats exhibit symptoms similar to those of obese patients with insulin-resistance or Type II diabetes; therefore, they have been used as a genetic model to study obesity, as well as a pharmacological model for the discovery of new drugs for the treatment of Type II diabetes and hyperlipidemia. In the present study, we compared the pharmacokinetics of two novel peroxisome proliferator-activated receptor (PPAR) agonists, MRL-I [(2R)-7-[3-[2-chloro-4-(4-fluorophenoxy)phenoxy]propoxy]-2-ethyl-3,4-dihydro-2H-benzopyran-2-carboxylic acid] and MRL-II [(2R)-7-[3-[2-chloro-4-(2,2,2-trifluoroethoxy)phenoxy]propoxy]-3,4-dihydro-2-methyl-2H-benzopyran-2-carboxylic acid], in obese Zucker and lean Sprague-Dawley rats following a single intravenous administration. The plasma clearance of both MRL-I and MRL-II was significantly lower in obese Zucker rats (4- and 2-fold, respectively) compared with Sprague-Dawley rats, but without any significant change in the volume of distribution, which resulted in a dramatic increase in the half-life (7- and 3-fold, respectively). The reversible in vitro plasma protein binding of [(14)C]MRL-I and [(14)C]MRL-II was comparable in the two strains, approximately 96% bound. The expression levels of uridine diphosphate-glucuronosyltransferases 1A1, 1A6, 2B1, and CYP2C11 and 3A1 mRNA in liver were lower (30-50%) in Zucker compared with Sprague-Dawley rats, as were the liver glutathione S-transferases (70%), quinone reductase (30%), organic anion-transporting protein 2 (80%), and multidrug resistance-associated protein 2 (Mrp2) (50%) mRNA levels. However, Mrp3 mRNA levels were similar in both strains. Consistent with these observations, the intrinsic clearance (CL(int)), calculated from the V(max)/K(m) of glucuronidation of [(14)C]MRL-I and [(14)C]MRL-II in liver microsomes, was approximately 2-fold lower in obese Zucker rats; the K(m) values were comparable in the two strains for both compounds. In conclusion, differences in the pharmacokinetics of two novel PPAR agonists, both cleared, predominantly, by conjugation, were evident in genetically obese Zucker rats compared with Sprague-Dawley rats. These differences were consistent with changes in the mRNA levels of hepatic drug-metabolizing enzymes and transporters. This information should be considered when comparing pharmacokinetic and efficacious doses in the obese Zucker rats, used as a pharmacological model, with those in Sprague-Dawley rats, which are used widely for drug metabolism and toxicology studies.


Subject(s)
Glucuronides/metabolism , Peroxisome Proliferator-Activated Receptors/pharmacokinetics , Rats, Sprague-Dawley/metabolism , Rats, Zucker/metabolism , Species Specificity , Animals , Benzopyrans/administration & dosage , Benzopyrans/blood , Benzopyrans/chemistry , Benzopyrans/metabolism , Benzopyrans/pharmacokinetics , Benzopyrans/pharmacology , Bile/chemistry , Bile/drug effects , Bile/metabolism , Blood Proteins/chemistry , Blood Proteins/drug effects , Blood Proteins/metabolism , Carbon Radioisotopes/administration & dosage , Carrier Proteins/metabolism , Disease Models, Animal , Drug Administration Schedule , Gene Expression/genetics , Glucuronides/chemistry , Glucuronosyltransferase/classification , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Half-Life , Injections, Intravenous , Male , Metabolic Clearance Rate , Microsomes, Liver/enzymology , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Protein Binding/drug effects , Protein Binding/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Zucker/genetics , Xenobiotics/metabolism
4.
J Pharmacol Exp Ther ; 311(1): 307-14, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15163679

ABSTRACT

Stimulation by quinidine of warfarin metabolism in vitro was first demonstrated with liver microsomal preparations. We report herein that this drug interaction is reproducible in an animal model but that it exhibits profound species differences. Thus, using rabbit liver microsomes and a kinetic model incorporating two binding sites, the hepatic intrinsic clearance of R-warfarin via the 10-hydroxylation pathway (CL(int)(W)) was projected to be 6 +/- 1 and 128 +/- 51 microl/min/g liver, respectively, in the absence and presence of 21 microM unbound quinidine. These estimates were consistent with the results from studies in which rabbit livers (n = 5) were perfused in situ with R-warfarin or R-warfarin plus quinidine. The CL(int)(W) increased from 7 +/- 3 to 156 +/- 106 microl/min/g liver after increasing the hepatic exposure of unbound quinidine from 0 to 21 microM. In contrast, when liver microsomes or intact livers from rats were examined, R-warfarin metabolism was inhibited by quinidine, the CL(int)(W) decreasing to 26% of the control value after exposure of perfused rat livers (n = 5) to 22 microM unbound quinidine. The third example involved monkey liver microsomes, in which the rate of 10-hydroxylation of R-warfarin was little affected in the presence of quinidine (<2-fold increase). In all three species, the 10-hydroxylation of R-warfarin was catalyzed primarily by members of CYP3A, based on immuno- and chemical inhibition analyses. These findings not only highlight the variability of drug interactions among different species but also suggest that changes in hepatic clearance resulting from stimulation of cytochrome P450 activity may be projected based on estimates generated from corresponding liver microsomal preparations.


Subject(s)
Microsomes, Liver/metabolism , Quinidine/pharmacology , Warfarin/metabolism , Animals , Drug Interactions , Hydroxylation/drug effects , Metabolic Clearance Rate , Muscarinic Antagonists/pharmacology , Perfusion , Rabbits , Rats , Species Specificity
5.
Drug Metab Dispos ; 32(2): 246-58, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14744948

ABSTRACT

The absorption, metabolism, and excretion of [14C]aprepitant, a potent and selective human substance P receptor antagonist for the treatment of chemotherapy-induced nausea and vomiting, was evaluated in rats and dogs. Aprepitant was metabolized extensively and no parent drug was detected in the urine of either species. The elimination of drug-related radioactivity, after i.v. or p.o. administration of [14C]aprepitant, was mainly via biliary excretion in rats and by way of both biliary and urinary excretion in dogs. Aprepitant was the major component in the plasma at the early time points (up to 8 h), and plasma metabolite profiles of aprepitant were qualitatively similar in rats and dogs. Several oxidative metabolites of aprepitant, derived from N-dealkylation, oxidation, and opening of the morpholine ring, were detected in the plasma. Glucuronidation represented an important pathway in the metabolism and excretion of aprepitant in rats and dogs. An acid-labile glucuronide of [14C]aprepitant accounted for approximately 18% of the oral dose in rat bile. The instability of this glucuronide, coupled with its presence in bile but absence in feces, suggested the potential for enterohepatic circulation of aprepitant via this conjugate. In dogs, the glucuronide of [14C]aprepitant, together with four glucuronides derived from phase I metabolites, were present as major metabolites in the bile, accounting collectively for approximately 14% of the radioactive dose over a 4- to 24-h period after i.v. dosing. Two very polar carboxylic acids, namely, 4-fluoro-alpha-hydroxybenzeneacetic acid and 4-fluoro-alpha-oxobenzeneacetic acid, were the predominant drug-related entities in rat and dog urine.


Subject(s)
Antiemetics/pharmacokinetics , Morpholines/pharmacokinetics , Neurokinin-1 Receptor Antagonists , Administration, Oral , Animals , Antiemetics/blood , Antiemetics/urine , Aprepitant , Bile/metabolism , Chromatography, High Pressure Liquid , Chromatography, Liquid , Dogs , Feces/chemistry , Glucuronides/blood , Glucuronides/urine , Injections, Intravenous , Liver/metabolism , Magnetic Resonance Spectroscopy , Male , Mandelic Acids/blood , Mandelic Acids/urine , Mass Spectrometry , Morpholines/blood , Morpholines/urine , Phenylacetates/blood , Phenylacetates/urine , Rats , Rats, Sprague-Dawley , Species Specificity
6.
Bioorg Med Chem Lett ; 13(2): 277-80, 2003 Jan 20.
Article in English | MEDLINE | ID: mdl-12482439

ABSTRACT

The development of potent, orally bioavailable (in rat) and selective dihydroquinazolinone inhibitors of p38alpha MAP kinase is described. These analogues are hybrids of a pyridinylimidazole p38alpha inhibitor reported by Merck Research Laboratories and VX-745. Optimization of the C-5 phenyl and the C-7 piperidinyl substituents led to the identification of 15i which gave excellent suppression of TNF-alpha production in LPS-stimulated whole blood (IC(50)=10nM) and good oral exposure in rats (F=68%, AUCn PO=0.58 microM h).


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Quinazolines/chemical synthesis , Quinazolines/pharmacology , Animals , Area Under Curve , Biological Availability , Drug Design , Enzyme Inhibitors/pharmacokinetics , In Vitro Techniques , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Molecular , Molecular Conformation , Monocytes/drug effects , Monocytes/metabolism , Rats , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases
7.
Drug Metab Dispos ; 30(7): 771-7, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12065435

ABSTRACT

The pharmacokinetics and oral bioavailability of (R)-N-[4-[2-[[2-hydroxy-2-(pyridin-3-yl)ethyl]amino]ethyl]phenyl]-4-[4-[4-(trifluoromethylphenyl]thiazol-2-yl]benzenesulfonamide (1), a 3-pyridyl thiazole benzenesulfonamide beta3-adrenergic receptor agonist, were investigated in rats, dogs, and monkeys. Systemic clearance was higher in rats (approximately 30 ml/min/kg) than in dogs and monkeys (both approximately 10 ml/min/kg), and oral bioavailability was 17, 27, and 4%, respectively. Since systemic clearance was 25 to 40% of hepatic blood flow in these species, hepatic extraction was expected to be low, and it was likely that oral bioavailability was limited either by absorption or a large first-pass effect in the gut. The absorption and excretion of 3H-labeled 1 were investigated in rats, and only 28% of the administered radioactivity was orally absorbed. Subsequently, the hepatic extraction of 1 was evaluated in rats (30%) and monkeys (47%). The low oral bioavailability in rats could be explained completely by poor oral absorption and hepatic first-pass metabolism; in monkeys, oral absorption was either less than in rats or first-pass extraction in the gut was greater. In an attempt to increase oral exposure, the pharmacokinetics and oral bioavailability of two potential prodrugs of 1, an N-ethyl [(R)-N-[4-[2-[ethyl[2-hydroxy-2-(3-pyridinyl)ethyl]amino]ethyl]phenyl]-4-[4-[4-(trifluoromethyl)phenyl]thiazol-2-yl]benzenesulfonamide; 2] and a morpholine derivative [(R)-N-[4-[2-[2-(3-pyridinyl)morpholin-4-yl]ethyl]phenyl]-4-[4-[4-(trifluoromethyl)- phenyl]thiazol-2-yl]benzenesulfonamide; 3], were evaluated in monkeys. Conversion to 1 was low (<3%) with both derivatives, and neither entity was an effective prodrug, but the oral bioavailability of 3 (56%) compared with 1 (4%) was significantly improved. The hypothesis that the increased oral bioavailability of 3 was due to a reduction in hydrogen bonding sites in the molecule led to the design of (R)-N-[4-[2-[[2-hydroxy-2-(pyridin-2-yl)ethyl]amino]ethyl]phenyl]-4-[4-(4-trifluoromethylphenyl)thiazol-2-yl]benzenesulfonamide (4), a 2-pyridyl beta3-adrenergic receptor agonist with improved oral bioavailability in rats and monkeys.


Subject(s)
Adrenergic beta-3 Receptor Agonists , Adrenergic beta-Agonists/pharmacokinetics , Sulfonamides/pharmacokinetics , Thiazoles/pharmacokinetics , Administration, Oral , Adrenergic beta-Agonists/chemistry , Adrenergic beta-Agonists/metabolism , Animals , Biological Availability , Dogs , Drug Evaluation, Preclinical , Macaca mulatta , Male , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-3/physiology , Sulfonamides/chemistry , Sulfonamides/metabolism , Thiazoles/chemistry , Thiazoles/metabolism , Benzenesulfonamides
8.
Drug Metab Dispos ; 30(7): 778-87, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12065436

ABSTRACT

(R)-N-[4-[2-[[2-Hydroxy-2-(pyridin-3-yl)ethyl]amino]ethyl]phenyl]- 4-[4-(4-trifluoro-methylphenyl)thiazol-2-yl]benzenesulfonamide (1) is a potent and selective agonist of the human beta3-adrenergic receptor. We report herein the data from studies of the metabolism and excretion of 1 in rats. Five metabolites were identified in the bile of male Sprague-Dawley rats administered 3H-labeled 1 by either oral gavage (10 mg/kg) or intravenous injection (3 mg/kg). These included a pyridine N-oxide derivative (M2), a primary amine resulting from N-dealkylation and loss of the pyridinyl-2-hydroxyethyl group (M4), a carboxylic acid derived from N-dealkylation and loss of the pyridyl-2-hydroxyethyl amine (M5), and the corresponding taurine and isethionic acid conjugates (M1 and M3). Metabolites M1 and M3 also were identified in rats treated with M5 and were generated in incubations of M5 with rat liver subcellular fractions in the presence of ATP and coenzyme A with supplementary taurine or isethionic acid. These results suggest that M5 is the precursor of M1 and M3 and that the formation of these conjugated metabolites follows similar mechanisms of amino acid conjugation. On the other hand, M2, M4, and M5 were produced from 1 in an NADPH-dependent manner in incubations with liver microsomes from rats, dogs, monkeys, and humans. In human liver preparations, these routes of biotransformation were shown to be catalyzed by cytochrome P450 3A4. In a bidirectional transport assay, transport of 1 across a monolayer of cells expressing P-glycoprotein (Pgp) was observed to be similar to that of vinblastine, which is an established substrate of the transporter protein. This finding, together with the observation that the parent compound was excreted in the feces of bile duct-cannulated animals following intravenous dosing, suggests that 1 is subject to Pgp-mediated excretion from intestine of rats.


Subject(s)
Adrenergic beta-3 Receptor Agonists , Isethionic Acid/metabolism , Sulfonamides/metabolism , Thiazoles/metabolism , Animals , Cell Line , Dogs , Female , Humans , Isethionic Acid/chemistry , Macaca mulatta , Male , Mice , Microsomes, Liver/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-3/metabolism , Sulfonamides/chemistry , Swine , Thiazoles/chemistry , Benzenesulfonamides
SELECTION OF CITATIONS
SEARCH DETAIL
...