Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Pharmacol ; 49(1): 80-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19004846

ABSTRACT

In this study, midazolam was used as a probe-sensitive CYP3A substrate to investigate the effect of anacetrapib on CYP3A activity, and ketoconazole was used as a probe-inhibitor to investigate the effect of potent CYP3A inhibition on the pharmacokinetics of anacetrapib, a novel cholesteryl ester transfer protein inhibitor in development for the treatment of dyslipidemia. Two partially blinded, randomized, 2-period, fixed-sequence studies were performed. Safety, tolerability, and midazolam and anacetrapib plasma concentrations were assessed. All treatments were generally well tolerated. The geometric mean ratios (90% confidence interval) of midazolam with anacetrapib/midazolam alone for AUC0-infinity and Cmax were 1.04 (0.94, 1.14) and 1.15 (0.97, 1.37), respectively. Exposure to anacetrapib was increased by ketoconazole--specifically, the geometric mean ratios (90% confidence interval) of anacetrapib with ketoconazole/anacetrapib alone for AUC0-infinity and Cmax were 4.58 (3.68, 5.71) and 2.37 (2.02, 2.78), respectively. The study showed that anacetrapib does not inhibit or induce CYP3A activity. Furthermore, anacetrapib appears to be a moderately sensitive substrate of CYP3A.


Subject(s)
Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cytochrome P-450 Enzyme System/physiology , Oxazolidinones/pharmacology , Adolescent , Adult , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Drug Interactions , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , In Vitro Techniques , Ketoconazole/pharmacology , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Midazolam/pharmacology , Middle Aged , Oxazolidinones/adverse effects , Young Adult
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.
J Pharmacol Exp Ther ; 304(3): 1161-71, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12604693

ABSTRACT

The mechanisms of pharmacokinetic interactions of a novel anti-human immunodeficiency virus (anti-HIV-1) antagonist of chemokine receptor 5 (CCR5) [2-(R)-[N-methyl-N-(1-(R)-3-(S)-((4-(3-benzyl-1-ethyl-(1H)-pyrazol-5-yl)piperidin-1-yl)methyl)-4-(S)-(3-fluorophenyl)cyclopent-1-yl)amino]-3-methylbutanoic acid (MRK-1)] with ritonavir were evaluated in rats and monkeys. MRK-1 was a good substrate for the human (MDR1) and mouse (Mdr1a) multidrug resistance protein transporters and was metabolized by CYP3A isozymes in rat, monkey, and human liver microsomes. Both the in vitro MDR1-mediated transport and oxidative metabolism of MRK-1 were inhibited by ritonavir. Although the systemic pharmacokinetics of MRK-1 in rats and monkeys were linear, the oral bioavailability increased with an increase in dose from 2 to 10 mg/kg. The area under the plasma concentration-time curve (AUC) of MRK-1 was increased 4- to 6-fold when a 2 or 10 mg/kg dose was orally coadministered with 10 mg/kg ritonavir. Further pharmacokinetic studies in rats indicated that P-glycoprotein (P-gp) inhibition by ritonavir increased the intestinal absorption of 2 mg/kg MRK-1 maximally by approximately 30 to 40%, and a major component of the interaction likely resulted from its reduced systemic clearance via the inhibition of CYP3A isozymes. Oral coadministration of quinidine (10 and 30 mg/kg) increased both the extent and the first-order rate of absorption of MRK-1 (2 mg/kg) by approximately 40 to 50% and approximately 100 to 300%, respectively, in rats, thus further substantiating the role of P-gp in modulating the intestinal absorption of MRK-1 in this species. At the 10 mg/kg MRK-1 dose, however, the entire increase in its AUC upon coadministration with ritonavir or quinidine could be attributed to a reduced systemic clearance, and no effects on intestinal absorption were apparent. In contrast to rats, the effects of P-gp in determining the intestinal absorption of MRK-1 appeared less significant in rhesus monkeys at either dose.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Aryl Hydrocarbon Hydroxylases/physiology , CCR5 Receptor Antagonists , Oxidoreductases, N-Demethylating/physiology , Pyrazoles/metabolism , Ritonavir/pharmacology , Valine/metabolism , Administration, Oral , Animals , Cytochrome P-450 CYP3A , Drug Interactions , HIV Protease Inhibitors/pharmacology , Haplorhini , Intestinal Absorption/drug effects , Male , Oxidation-Reduction , Protein Binding , Pyrazoles/pharmacokinetics , Quinidine/pharmacology , Rats , Rats, Sprague-Dawley , Valine/analogs & derivatives , Valine/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...