Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Drug Metab Pharmacokinet ; 42(5): 827-834, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28185143

ABSTRACT

BACKGROUND AND OBJECTIVES: 1-Aminobenzotriazole, a known time-dependent inhibitor of cytochrome P450 (CYP) enzymes, and ketoconazole, a strong inhibitor of the human CYP3A4 isozyme, are used as standard probe inhibitors to characterize the CYP and/or non-CYP-mediated metabolism of xenobiotics. In the present investigation, 1-Aminobenzotriazole and ketoconazole are characterized as potent monoamine oxidase (MAO) inhibitors in vitro using mouse, rat and human liver microsomes and S9 fractions. METHODS: Inhibition potential of 1-aminobenzotriazole and ketoconazole was studied in mice, rat and human liver microsomes, S9 fractions, MAO-A and MAO-B expressed enzymes by monitoring the formation of 4-hydroxyquinoline (4-HQ) from kynuramine, a specific substrate of MAO by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Mechanism of MAO inhibition was studied by incubating varying concentration of kynuramine with mouse, rat and human S9 fractions at varying concentration of 1-aminobenzatriazole and ketoconazole and monitoring the formation of 4-HQ. RESULTS: 1-aminobenzotriazole and ketoconazole inhibited both MAO isozymes (MAO-A and MAO-B) with more specificity towards MAO-B. Kynuramine substrate kinetics in mouse, rat and human S9 fractions with varying 1-aminobenzotriazole and ketoconazole concentrations showed decreased maximum rate (V max) for 4-HQ formation without affecting the Michaelis-Menten constant (K m). A non-competitive inhibition model was constructed and inhibition constants (K i) for 1-aminobenzotriazole (7.87 ± 0.61, 8.61 ± 0.92, 65.2 ± 1.61 µM for mice, rat and humans, respectively) and ketoconazole (0.12 ± 0.01, 2.04 ± 0.08, 5.52 ± 0.47 µM for mice, rat and humans, respectively) were determined. CONCLUSIONS: 1-Aminobenzotriazole and ketoconazole are characterized as non-competitive inhibitors of mice, rat and human MAO in vitro and the extent of their MAO inhibition potential is species specific. 1-Aminobenzotriazole or ketoconazole can be used as a probe inhibitor in vitro for screening the involvement of MAO-dependent metabolism of new chemical entities (NCE) in early drug discovery.


Subject(s)
Ketoconazole/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Triazoles/pharmacology , Animals , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme Inhibitors/metabolism , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/metabolism , Ketoconazole/metabolism , Kinetics , Liver/metabolism , Mice , Microsomes, Liver/metabolism , Monoamine Oxidase Inhibitors/metabolism , Rats , Triazoles/metabolism
2.
J Pharm Sci ; 105(6): 1976-1986, 2016 06.
Article in English | MEDLINE | ID: mdl-27103011

ABSTRACT

The anticoagulant drug warfarin and the lipid-lowering statin drugs are commonly co-administered to patients with cardiovascular diseases. Clinically significant drug-drug interactions (DDIs) between these drugs have been recognized through case studies for many years, but the biochemical mechanisms causing these interactions have not been explained fully. Previous theories include kinetic alterations in cytochrome P-450-mediated drug metabolism or disturbances of drug-protein binding, leading to anticoagulant activity of warfarin; however, neither the enantioselective effects on warfarin metabolism nor the potential disruption of drug transporter function have been well investigated. This study investigated the etiology of the DDIs between warfarin and statins. Liquid chromatography-mass spectrometry methods were developed and validated to quantify racemic warfarin, 6 of its hydroxylated metabolites, and pure enantiomers of warfarin; these methods were applied to study the role of different absorption, distribution, metabolism, and excretion properties, leading to DDIs. Plasma protein binding displacement of warfarin was performed in the presence of statins using equilibrium dialysis method. Substrate kinetics of warfarin and pure enantiomers were performed with human liver microsomes to determine the kinetic parameters (Km and Vmax) for the formation of all 6 hydroxywarfarin metabolites, inhibition of warfarin metabolism in the presence of statins, was determined. Uptake transport studies of warfarin were performed using overexpressing HEK cell lines and efflux transport using human adenocarcinoma colonic cell line cells. Fluvastatin significantly displaced plasma protein binding of warfarin and pure enantiomers; no other statin resulted in significant displacement of warfarin. All the statins that inhibited the formation of 10-hydroxywarfarin, atorvastatin, pitavastatin, and simvastatin were highly potent compared to other statins; in contrast, only fluvastatin was found to be a potent inhibitor of formation of 7-hydroxy warfarin. Uptake and efflux drug transporters do not play any role in these DDIs. The results showed that DDIs between warfarin and statins are primarily caused by cytochrome P-450 inhibition.


Subject(s)
Anticoagulants/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Warfarin/metabolism , Caco-2 Cells , Dose-Response Relationship, Drug , Drug Interactions/physiology , HEK293 Cells , Humans , Protein Binding
3.
Article in English | MEDLINE | ID: mdl-26655108

ABSTRACT

Warfarin is an anticoagulant used in the treatment of thrombosis and thromboembolism. It is given as a racemic mixture of R and S enantiomers. These two enantiomers show differences in metabolism by CYPs: S-warfarin undergoes 7 hydroxylation by CYP2C9 and R-warfarin by CYP3A4 to form 10 hydroxy warfarin. In addition, warfarin is acted upon by different CYPs to form the minor metabolites 3'-hydroxy, 4'-hydroxy, 6-hydroxy, and 8-hydroxy warfarin. For analysis, separation of these metabolites is necessary since all have the same m/z ratio and similar fragmentation pattern. Enzyme kinetics for the formation of all of the six hydroxylated metabolites of warfarin from human liver microsomes were determined using an LC-MS/MS QTrap and LC-MS/MS with a differential mobility spectrometry (DMS) (SelexION™) interface to compare the kinetic parameters. These two methods were chosen to compare their selectivity and sensitivity. Substrate curves for 3'-OH, 4'-OH, 6-OH, 7-OH, 8-OH and 10-OH warfarin formation were generated to determine the kinetic parameters (Km and Vmax) in human liver microsomal preparations. The limit of quantitation (LOQ) for all the six hydroxylated metabolites of warfarin were in the range of 1-3nM using an LC-MS/MS QTrap method which had a run time of 22min. In contrast, the LOQ for all the six hydroxylated metabolites using DMS interface technology was 100nM with a run time of 2.8min. We compare these two MS methods and discuss the kinetics of metabolite formation for the metabolites generated from racemic warfarin. In addition, we show inhibition of major metabolic pathways of warfarin by sulfaphenazole and ketoconazole which are known specific inhibitors of CYP2C9 and CYP3A4 respectively.


Subject(s)
Anticoagulants/pharmacokinetics , Chromatography, Liquid/methods , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP3A/metabolism , Tandem Mass Spectrometry/methods , Warfarin/pharmacokinetics , Calibration , Humans , Reproducibility of Results
4.
Drug Metab Dispos ; 39(4): 693-702, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21177487

ABSTRACT

Oxymetazoline (6-tert-butyl-3-(2-imidazolin-2-ylmethyl)-2,4-dimethylphenol) has been widely used as a nonprescription nasal vasoconstrictor for >40 years; however, its metabolic pathway has not been investigated. This study describes the in vitro metabolism of oxymetazoline in human, rat, and rabbit liver postmitochondrial supernatant fraction from homogenized tissue (S9) fractions and their microsomes supplemented with NADPH. The metabolites of oxymetazoline identified by liquid chromatography (LC)/UV/tandem mass spectrometry (MS/MS), included M1 (monohydroxylation of the t-butyl group), M2 (oxidative dehydrogenation of the imidazoline to an imidazole moiety), M3 (monohydroxylation of M2), M4 (dihydroxylation of oxymetazoline), and M5 (dihydroxylation of M2). Screening with nine human expressed cytochromes P450 (P450s) identified CYP2C19 as the single P450 isoform catalyzing the formation of M1, M2, and M3. Glutathione conjugates of oxymetazoline (M6) and M2 (M7) were identified in the liver S9 fractions, indicating the capability of oxymetazoline to undergo bioactivation to reactive intermediate species. M6 and M7 were not detected in those liver S9 incubations without NADPH. Cysteine conjugates (M8 and M9) derived from glutathione conjugates and hydroxylated glutathione conjugates (M10 and M11) were also identified. The reactive intermediate of oxymetazoline was trapped with glutathione and N-acetyl cysteine and identified by LC/MS/MS. M6 was isolated and identified by one-dimensional or two-dimensional NMR as the glutathione conjugate of a p-quinone methide. We have shown the tendency of oxymetazoline to form p-quinone methide species via a bioactivation mechanism involving a CYP2C19-catalyzed two-electron oxidation. Nevertheless, we conclude that the formation of this reactive species might not be a safety concern for oxymetazoline nasal products because of the typical low-dose and brief dosage regimen limited to nasal delivery.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Glutathione/metabolism , Oxymetazoline/metabolism , Sympathomimetics/metabolism , Acetylcysteine/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 CYP2C19 , Humans , Hydroxylation , In Vitro Techniques , Indolequinones/metabolism , Liver/metabolism , Male , Microsomes, Liver/metabolism , NADP/metabolism , Oxidation-Reduction , Oxymetazoline/chemistry , Rabbits , Rats , Sympathomimetics/chemistry
5.
Pharmacology ; 73(3): 113-20, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15499226

ABSTRACT

The purpose of this study was to determine whether inhibition of potassium channels or cytochrome P450 attenuates the transient phase of hypotension during endotoxic shock in vivo, and to determine whether these interventions improve the rate of survival. Male Sprague-Dawley rats were pretreated with saline (0.2 ml, i.v.), tetraethylammonium chloride (TEA 30 mg/kg; 0.2 ml, i.v.), proadifen (SKF-525 A; 50 mg/kg, i.p.) or ketoconazole (50 mg/kg, i.p.) and challenged with lipopolysaccharide (LPS; 20 mg/kg, i.p.). Changes in heart rate, mean (MAP), systolic (SP) and diastolic (DP) arterial pressures as well as survival rate were then monitored for 45 min. Potassium channel inhibition with TEA had no effect on LPS-induced hypotension at any time point compared with saline (maximal fall in MAP of 79 +/- 18 and 80 +/- 13 mm Hg, respectively). Pretreatment with proadifen or ketoconazole, inhibitors of cytochrome P450, significantly attenuated LPS-induced hypotension compared with saline (maximal fall in MAP of 34, 26 and 63% below baseline, respectively). This effect was evident in all arterial pressures measured, MAP, SP and DP. At 45 min, the survival rate in the saline group was 66%. Pretreatment with TEA significantly reduced survival rate to 50% and pretreatment with proadifen or ketoconazole improved survival to 100% (p < 0.05). These results suggest that an arachidonic acid metabolite produced by a cytochrome P450-catalyzed reaction may contribute to the transient phase of LPS-induced hypotension. However, these effects do not appear to be mediated through potassium channel activation.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Hypotension/metabolism , Lipopolysaccharides/toxicity , Potassium Channel Blockers/pharmacology , Potassium Channels/physiology , Shock, Septic/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Hypotension/chemically induced , Hypotension/drug therapy , Male , Potassium Channel Blockers/therapeutic use , Potassium Channels/metabolism , Rats , Rats, Sprague-Dawley , Shock, Septic/chemically induced , Shock, Septic/drug therapy
6.
Clin Pharmacol Ther ; 74(2): 121-9, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12891222

ABSTRACT

BACKGROUND: Components of grapefruit juice may impair the activity of intestinal cytochrome P450 (CYP) 3A enzymes, sometimes resulting in clinically important drug interactions. The time course of recovery from CYP3A inhibition after a single exposure to grapefruit juice is not clearly established. METHODS: Healthy volunteer subjects (N = 25) received a single 6-mg oral dose of the CYP3A substrate midazolam in the control condition without exposure to grapefruit juice. Two days later, midazolam was administered 2 hours after 300 mL of regular-strength grapefruit juice. Subjects were then randomly assigned to 3 different groups, receiving a third midazolam challenge at 26, 50, or 74 hours after exposure to grapefruit juice. The capacity of 6'7'-dihydroxybergamottin and bergamottin to inhibit human CYP3A was studied in vitro using human liver microsomes. RESULTS: The area under the plasma concentration curve (AUC) for midazolam increased by a factor of 1.65 (ratio compared with control) when midazolam was given 2 hours after grapefruit juice. At 26, 50, and 74 hours after grapefruit juice, the AUC ratios (mean AUC value at the indicated time divided by the mean control AUC on day 1) were 1.29, 1.29, and 1.06, respectively. The relationship of time after grapefruit juice exposure versus AUC increase over control indicated a recovery half-life estimated at 23 hours. The midazolam elimination half-life did not change significantly from the control value at any time after grapefruit juice exposure. 6'7'-Dihydroxybergamottin inhibited midazolam alpha-hydroxylation in vitro, with a mean 50% inhibitory concentration of 4.7 micro mol/L; preincubation of microsomes with 6'7'-dihydroxybergamottin greatly reduced the 50% inhibitory concentration to 0.31 micro mol/L, consistent with mechanism-based inhibition. Bergamottin itself had much weaker inhibitory potency compared to 6'7'-dihydroxybergamottin. CONCLUSIONS: A usual single exposure to grapefruit juice appears to impair the enteric, but not the hepatic, component of presystemic extraction of oral midazolam. Recovery is largely complete within 3 days, consistent with enzyme regeneration after mechanism-based inhibition. 6'7'-Dihydroxybergamottin was verified as a potent mechanism-based inhibitor of midazolam alpha-hydroxylation by CYP3A in vitro.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Beverages , Citrus paradisi , Oxidoreductases, N-Demethylating/metabolism , Adult , Area Under Curve , Biotransformation , Chromatography, Gas , Cytochrome P-450 CYP3A , Female , Furocoumarins/pharmacology , Half-Life , Humans , Hypnotics and Sedatives/pharmacokinetics , Isoenzymes/metabolism , Kinetics , Liver/metabolism , Male , Midazolam/pharmacokinetics , Middle Aged , Triazolam/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...