Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Ther Drug Monit ; 30(3): 276-81, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18520598

ABSTRACT

Previous studies reported omeprazole to be an inhibitor of cytochrome P450 (CYP) 2C19 and suggested the pharmacokinetic interaction of omeprazole with R-warfarin. The aim of this study was to compare possible effects of omeprazole on the stereoselective pharmacokinetics and pharmacodynamics of warfarin between CYP2C19 genotypes. Seventeen subjects, of whom 10 were homozygous extensive metabolizers (hmEMs) and seven were poor metabolizers (PMs) for CYP2C19, were enrolled in this randomized crossover study, and they ingested 20 mg omeprazole or placebo once daily for 11 days. On day 7, they administered a single dose of 10 mg racemic warfarin. The plasma concentrations of warfarin enantiomers and prothrombin time expressed as international normalized ratio were monitored up to 120 hours. During the placebo phase, area under the plasma concentration-time curve (AUC) and elimination half-life (t1/2) of R-warfarin in PMs was significantly greater than those in hmEMs (AUC[0-infinity], 42,938/34,613 ng h/mL [PM/hmEM], P = 0.004; t1/2, 48.8/40.8 hours [PM/hmEM], P = 0.013). Omeprazole treatment significantly increased the AUC(0-infinity) (41,387 ng h/mL, P = 0.004) and t1/2 (46.4 hours, P = 0.017) of R-warfarin in hmEMs to levels comparable to those in the PMs. There were no differences in S-warfarin pharmacokinetics between the CYP2C19 genotypes (AUC[0-infinity], 15,851/16,968 ng*h/mL [PM/hmEM]; t1/2, 22.7/25.4 h [PM/hmEM]), or between the two treatment phases (AUC[0-infinity], 14,756/18,166 ng h/mL [PM/hmEM]; t1/2, 27.0/25.4 hours [PM/hmEM] in the omeprazole phase) as well as anticoagulant effects. These results indicate that CYP2C19 activity was one of determinants on the R-warfarin disposition because the pharmacokinetics of warfarin enantiomers were different between the CYP2C19 genotypes and the omeprazole affected the R-warfarin pharmacokinetics of CYP2C19 in only hmEMs. However, the phamacodynamic effect of the interaction of warfarin with omeprazole would be of minor clinical significance.


Subject(s)
Anticoagulants/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Omeprazole/pharmacokinetics , Proton Pump Inhibitors/pharmacokinetics , Warfarin/pharmacokinetics , Adult , Alleles , Area Under Curve , Biotransformation , Cytochrome P-450 CYP2C19 , Double-Blind Method , Drug Interactions , Female , Genotype , Humans , Male , Phenotype , Prothrombin Time , Stereoisomerism
2.
Eur J Clin Pharmacol ; 63(11): 1031-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17701405

ABSTRACT

OBJECTIVE: The purpose of this study was to identify the common time point to achieve hydroxylation index (HI: omeprazole plasma concentration/5-hydroxyomeprazole plasma concentration) reflecting AUCOPZ/AUC5OH-OPZ after intravenous (IV) and oral (PO) administration. METHODS: Twenty young and 28 elderly healthy subjects, including different CYP2C19 genotypes, were enrolled in the study. The young subjects received either 40 mg PO or 20 mg IV omeprazole, whereas the elderly subjects received 10 mg IV. The relation between AUCOPZ/AUC5OH-OPZ and HI was determined by Spearman's rank correlation. Multiple stepwise linear regression analysis was performed to identify the common time point to calculate HI that reflects AUCOPZ/AUC5OH-OPZ after IV. RESULTS: In the correlation between HI and AUCOPZ/AUC5OH-OPZ IV at observed time points, HI3h showed the highest correlation coefficients (r = 0.894, p < 0.001) in all 48 subjects. The correlation of HI between IV and PO at observed time points showed that HI3h was highest (r = 0.916, p < 0.001) in 20 young subjects. Additionally, there was no significant difference between HI(3h) of IV and that of PO (12.9 +/- 15.9 and 12.9 +/- 15.1, p = 0.997). The regression equation of HI3h was the best to estimate AUCOPZ/AUC5OH-OPZ (AUCOPZ/AUC5OH-OPZ = 1.37 * HI3h + 0.18 * Age - 7.83, r2 = 0.883, p < 0.001). CONCLUSIONS: This study demonstrated that HI3h after omeprazole IV was able to estimate AUCOPZ/AUC5OH-OPZ, as well as HI3h after PO. Additionally, CYP2C19 activity can be estimated more definitely by using HI after omeprazole IV without intestinal absorption.


Subject(s)
2-Pyridinylmethylsulfinylbenzimidazoles/pharmacokinetics , Anti-Ulcer Agents/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/metabolism , Drug Monitoring/methods , Mixed Function Oxygenases/metabolism , Omeprazole/pharmacokinetics , Administration, Oral , Adult , Age Factors , Aged , Aged, 80 and over , Anti-Ulcer Agents/administration & dosage , Area Under Curve , Cross-Over Studies , Cytochrome P-450 CYP2C19 , Female , Genotype , Humans , Hydroxylation , Injections, Intravenous , Linear Models , Male , Omeprazole/administration & dosage , Time Factors
3.
Ther Drug Monit ; 29(3): 333-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17529891

ABSTRACT

A simple and sensitive column-switching high-performance liquid chromatographic method for the simultaneous determination of warfarin enantiomers and their metabolites, 7-hydroxywarfarin enantiomers, in human plasma is described. Warfarin enantiomers, 7-hydroxywarfarin enantiomers, and an internal standard, diclofenac sodium, were extracted from 1 mL of a plasma sample using diethyl ether-chloroform (80:20, v/v). The extract was injected onto column I (TSK precolumn BSA-C8, 5 microm, 10 mm x 4.6 mm inside diameter) for cleanup and column II (Chiralcel OD-RH analytical column, 150 mm x 4.6 mm inside diameter) coupled with a guard column (Chiralcel OD-RH guard column, 10 mm x4.6 mm inside diameter) for separation. The mobile phase consisted of phosphate buffer-acetonitrile (84:16 v/v, pH 2.0) for clean-up and phosphate buffer-acetonitrile (45:55 v/v, pH 2.0) for separation. The peaks were monitored with an ultraviolet detector set at a wavelength of 312 nm, and total time for chromatographic separation was approximately 25 minutes. The validated concentration ranges of this method were 3 to 1000 ng/mL for (R)- and (S)-warfarin and 3 to 200 ng/mL for (R)- and (S)-7-hydroxywarfarin. Intra- and interday coefficients of variation were less than 4.4% and 4.9% for (R)-warfarin and 4.8% and 4.0% for (S)-warfarin, and 5.1% and 4.2% for (R)-7-hydroxywarfarin and 5.8% and 5.0% for (S)-7-hydroxywarfarin at the different concentrations. The limit of quantification was 3 ng/mL for both warfarin and 7-hydroxywarfarin enantiomers. This method was suitable for therapeutic drug monitoring of warfarin enantiomers and was applied in a pharmacokinetic study requiring the simultaneous determination of warfarin enantiomers and its metabolite, 7-hydroxywarfarin enantiomers, in human volunteers.


Subject(s)
Anticoagulants/metabolism , Chromatography, High Pressure Liquid/methods , Drug Monitoring/methods , Warfarin/analogs & derivatives , Warfarin/metabolism , Anticoagulants/blood , Humans , Stereoisomerism , Warfarin/blood
4.
Eur J Clin Pharmacol ; 63(2): 143-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17203292

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the absolute bioavailability and the metabolism of omeprazole following single intravenous and oral administrations to healthy subjects in relation to CYP2C19 genotypes. METHODS: Twenty subjects, of whom 6 were homozygous extensive metabolizers (hmEMs), 8 were heterozygous EMs (htEMs) and 6 were poor metabolizers (PMs) for CYP2C19, were enrolled in this study. Each subject received either a single omeprazole 20 mg intravenous dose (IV) or 40 mg oral dose (PO) in a randomized fashion during 2 different phases. RESULTS: Mean omeprazole AUC (0,infinity) was 1164, 3093 and 10511 ng h/mL after PO, and 1435, 2495 and 6222 ng h/mL after IV in hmEMs, htEMs and PMs, respectively. Therefore, the absolute bioavailability of omeprazole in PMs was significantly higher than that in hmEMs (p < 0.001) and htEMs (p < 0.001). Hydroxylation metabolic indexes after IV and PO were significantly lower in PMs than in hmEMs (p < 0.001) and htEMs (p < 0.001), and was correlated with the absolute bioavailability (p < 0.0001 for both IV and PO). Sulfoxidation metabolic index after IV was significantly different between the CYP2C19 genotypes, whereas no difference was found after a single oral dose. CONCLUSION: This study indicates that the absolute bioavailability of omeprazole differs among the three different CYP2C19 genotypes after a single dose of omeprazole orally or intravenously. Hydroxylation metabolic index of omeprazole may be mainly attributable to the genotype of CYP2C19. As for the sulfoxidation metabolic index after a single oral dose, intestinal CYP3A may be contributed to omeprazole metabolism.


Subject(s)
Anti-Ulcer Agents/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/genetics , Mixed Function Oxygenases/genetics , Omeprazole/pharmacokinetics , Administration, Oral , Adult , Anti-Ulcer Agents/administration & dosage , Area Under Curve , Biological Availability , Cross-Over Studies , Cytochrome P-450 CYP2C19 , Dose-Response Relationship, Drug , Female , Heterozygote , Homozygote , Humans , Hydroxylation , Injections, Intravenous , Male , Omeprazole/administration & dosage , Oxidation-Reduction
5.
Cancer Chemother Pharmacol ; 59(5): 575-80, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16912889

ABSTRACT

PURPOSE: The aim of this study was to determine the pharmacokinetics of low-dose nedaplatin combined with paclitaxel and radiation therapy in patients having non-small-cell lung carcinoma and establish the optimal dosage regimen for low-dose nedaplatin. We also evaluated predictive accuracy of reported formulas to estimate the area under the plasma concentration-time curve (AUC) of low-dose nedaplatin. PATIENTS AND METHODS: A total of 19 patients were administered a constant intravenous infusion of 20 mg/m(2) body surface area (BSA) nedaplatin for an hour, and blood samples were collected at 1, 2, 3, 4, 6, 8, and 19 h after the administration. Plasma concentrations of unbound platinum were measured, and the actual value of platinum AUC (actual AUC) was calculated based on these data. The predicted value of platinum AUC (predicted AUC) was determined by three predictive methods reported in previous studies, consisting of Bayesian method, limited sampling strategies with plasma concentration at a single time point, and simple formula method (SFM) without measured plasma concentration. Three error indices, mean prediction error (ME, measure of bias), mean absolute error (MAE, measure of accuracy), and root mean squared prediction error (RMSE, measure of precision), were obtained from the difference between the actual and the predicted AUC, to compare the accuracy between the three predictive methods. RESULTS: The AUC showed more than threefold inter-patient variation, and there was a favorable correlation between nedaplatin clearance and creatinine clearance (Ccr) (r = 0.832, P < 0.01). In three error indices, MAE and RMSE showed significant difference between the three AUC predictive methods, and the method of SFM had the most favorable results, in which %ME, %MAE, and %RMSE were 5.5, 10.7, and 15.4, respectively. CONCLUSIONS: The dosage regimen of low-dose nedaplatin should be established based on Ccr rather than on BSA. Since prediction accuracy of SFM, which did not require measured plasma concentration, was most favorable among the three methods evaluated in this study, SFM could be the most practical method to predict AUC of low-dose nedaplatin in a clinical situation judging from its high accuracy in predicting AUC without measured plasma concentration.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Organoplatinum Compounds/pharmacokinetics , Aged , Algorithms , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Area Under Curve , Body Surface Area , Carcinoma, Non-Small-Cell Lung/drug therapy , Data Interpretation, Statistical , Female , Half-Life , Humans , Infusions, Intravenous , Lung Neoplasms/drug therapy , Male , Middle Aged , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/therapeutic use , Predictive Value of Tests , Reproducibility of Results
6.
Article in English | MEDLINE | ID: mdl-17085083

ABSTRACT

This paper describes sensitive and reliable determination of midazolam (MDZ) and its major metabolite 1'-hydroxymidazolam (1-OHMDZ) in human plasma by liquid chromatography-mass spectrometry (LC-MS) with a sonic spray ionization (SSI) interface. MDZ, 1-OHMDZ and diazepam as an internal standard were extracted from 1ml of alkalinized plasma using n-hexane-chloroform (70:30, v/v). The extract was injected into an analytical column (YMC-Pak Pro C(18), 50mmx2.0mmi.d.). The mobile phase for separation consisted of 10mM ammonium acetate and methanol (50:50, v/v) and was delivered at a flow-rate of 0.2ml/min. The drift voltage was 100V. The sampling aperture was heated at 120 degrees C and the shield temperature was 260 degrees C. The total time for chromatographic separation was less than 16min. The validated concentration ranges of this method were 0.25-50ng/ml for both MDZ and 1-OHMDZ. Mean recoveries were 93.6% for MDZ and 86.6% for 1-OHMDZ. Intra- and inter-day coefficient variations were less than 6.5 and 5.5% for MDZ, and 6.1 and 5.7% for 1-OHMDZ at 0.3, 4, 20 and 40ng/ml. The limits of quantification were 0.25ng/ml for both MDZ and 1-OHMDZ. This method was sensitive and reliable enough for pharmacokinetic studies on healthy volunteers, and was applied for the measurement of CYP3A activity in humans after an intravenous (1mg) and a single-oral administration (2mg) of subtherapeutic MDZ dose.


Subject(s)
Chromatography, Liquid/methods , Cytochrome P-450 CYP3A/metabolism , Mass Spectrometry/methods , Midazolam/analogs & derivatives , Midazolam/blood , Adult , Female , Humans , Male , Midazolam/chemistry , Midazolam/metabolism , Midazolam/pharmacokinetics , Molecular Structure , Reproducibility of Results
7.
Eur J Clin Pharmacol ; 62(10): 855-61, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16915367

ABSTRACT

OBJECTIVE: The purpose of this study is to evaluate whether a simple formula using limited blood samples can predict the area under the plasma rabeprazole concentration-time curve (AUC) in co-administration with CYP inhibitors. METHODS: A randomized double-blind placebo-controlled crossover study design in three phases was conducted at intervals of 2 weeks. Twenty-one healthy Japanese volunteers, including three CYP2C19 genotype groups, took a single oral 20-mg dose of rabeprazole after three 6-day pretreatments, i.e., clarithromycin 800 mg/day, fluvoxamine 50 mg/day, and placebo. Prediction formulas of the AUC were derived from pharmacokinetics data of 21 subjects in three phases using multiple linear regression analysis. Ten blood samples were collected over 24 h to calculate AUC. Plasma concentrations of rabeprazole was measured by an HPLC-assay (l.l.q.=1 ng/ml). RESULTS: The AUC was based on all the data sets (n=63). The linear regression using two points (C3 and C6) could predict AUC(0-infinity) precisely, irrespective of CYP2C19 genotypes and CYP inhibitors (AUC(0-infinity)=1.39xC3+7.17xC6+344.14, r (2)=0.825, p<0.001). CONCLUSION: The present study demonstrated that the AUC of rabeprazole can be estimated by the simple formula using two-point concentrations. This formula can be more accurate for the prediction of AUC estimation than that reflected by CYP2C19 genotypes without any determination, even if there are significant differences for the CYP2C19 genotypes. Therefore, this prediction formula might be useful to evaluate whether CYP2C19 genotypes really reflects the curative effect of rabeprazole.


Subject(s)
2-Pyridinylmethylsulfinylbenzimidazoles/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , 2-Pyridinylmethylsulfinylbenzimidazoles/blood , 2-Pyridinylmethylsulfinylbenzimidazoles/metabolism , Administration, Oral , Adult , Algorithms , Area Under Curve , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Chromatography, High Pressure Liquid , Clarithromycin/administration & dosage , Clarithromycin/blood , Clarithromycin/pharmacokinetics , Cross-Over Studies , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Double-Blind Method , Enzyme Inhibitors/metabolism , Female , Fluvoxamine/administration & dosage , Fluvoxamine/blood , Fluvoxamine/pharmacokinetics , Half-Life , Heterozygote , Homozygote , Humans , Male , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Rabeprazole , Regression Analysis , Sulfides/blood , Sulfides/metabolism , Time Factors
8.
Ther Drug Monit ; 28(4): 526-31, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16885720

ABSTRACT

A simple and sensitive column-switching high-performance liquid chromatographic method for the simultaneous determination of itraconazole (ITZ) and its active metabolite, hydroxyitraconazole (HIT) in human plasma is described. ITZ, HIT, and an internal standard, R051012, were extracted from 1 mL of alkalinized plasma sample using n-heptane-chloroform (60:40, vol/vol). The extract was injected onto column I (TSK precolumn BSA-ODS/S, 5 microm, 10 x 4.6 mm ID) for clean-up and column II (Develosil C8-5 column, 5 microm, 150 x 4.6 mm ID) for separation. The mobile phase consisted of phosphate buffer-acetonitrile (68:32 vol/vol, pH 6.0) for clean-up and phosphate buffer-acetonitrile (35:65 vol/vol, pH 6.0) for separation. The peaks were monitored with an ultraviolet detector set at a wavelength of 263 nm, and total time for chromatographic separation was about 24 minutes. The validated concentration ranges of this method were 3 to 500 ng/mL for ITZ and 3 to 1000 ng/mL for HIT. Mean recoveries were 59.7% for ITZ and 72.8% for HIT. Intraday and interday coefficients of variation were less than 4.6% and 5.0% for ITZ, and 4.6% and 4.9% for HIT at the different concentrations. The limit of quantification was 3 ng/mL for both ITZ and HIT. This method was suitable for therapeutic drug monitoring of ITZ and HIT, and was applied to pharmacokinetic studies in human volunteers.


Subject(s)
Chromatography, High Pressure Liquid/methods , Itraconazole/analogs & derivatives , Itraconazole/blood , Spectrophotometry, Ultraviolet/methods , Adult , Antifungal Agents/blood , Antifungal Agents/metabolism , Antifungal Agents/pharmacokinetics , Area Under Curve , Chloroform/chemistry , Chromatography, High Pressure Liquid/instrumentation , Chromatography, High Pressure Liquid/standards , Drug Monitoring/methods , Drug Stability , Fasting/blood , Heptanes/chemistry , Humans , Itraconazole/metabolism , Itraconazole/pharmacokinetics , Male , Metabolic Clearance Rate , Reproducibility of Results , Spectrophotometry, Ultraviolet/standards , Time Factors
9.
Drug Metab Dispos ; 34(11): 1875-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16928786

ABSTRACT

The aim of this study was to determine the inhibitory effect of itraconazole at different coadministered doses on fexofenadine pharmacokinetics. In a randomized four-phase crossover study, 11 healthy volunteers were administered a 60-mg fexofenadine hydrochloride tablet alone on one occasion (control phase) and with three different doses of 50, 100, and 200 mg of itraconazole simultaneously on the other three occasions (itraconazole phase). Although the elimination half-life and the renal clearance of fexofenadine remained relatively constant, a single administration of itraconazole with fexofenadine significantly increased mean area under the plasma concentration-time curve (AUC(0-infinity)) of fexofenadine (1701/3554, 4308, and 4107 ng h/ml for control; 50 mg, 100 mg, and 200 mg of itraconazole, respectively). Although mean itraconazole AUC(0-48) from 50 mg to 200 mg increased dose dependently from 214 to 772 ng h/ml (p = 0.003), no significant difference was noted in the three parameters, AUC (p = 0.423), C(max) (p = 0.636), and renal clearance (p = 0.495), of fexofenadine among the three doses of itraconazole. Itraconazole exposure at a lower dose (50 mg) compared with the clinical dose (200 mg once or twice daily) had the maximal effect on fexofenadine pharmacokinetics, even though itraconazole plasma concentrations gradually increased after higher doses. These findings suggest that the interaction may occur at the gut wall before reaching the portal vein circulation, and the inhibitory effect must be saturated by substantial local concentrations of itraconazole in the gut lumen after 50-mg dosing.


Subject(s)
Antifungal Agents/pharmacology , Histamine H1 Antagonists, Non-Sedating/pharmacokinetics , Itraconazole/pharmacology , Terfenadine/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Administration, Oral , Adult , Aged , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Interactions , Female , Histamine H1 Antagonists, Non-Sedating/blood , Histamine H1 Antagonists, Non-Sedating/urine , Humans , Male , Middle Aged , Terfenadine/blood , Terfenadine/pharmacokinetics , Terfenadine/urine
10.
Br J Clin Pharmacol ; 62(3): 372-6, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16796706

ABSTRACT

AIMS: We determined whether or not the extent of drug interaction of fexofenadine by itraconazole is time-dependent. METHODS: In a randomized two-phase crossover study, itraconazole was administered orally for 6 days, and, on days 1, 3 and 6, fexofenadine was administered simultaneously. On another occasion, fexofenadine was administered alone. RESULTS: Itraconazole increased fexofenadine AUC(0, infinity), and the % change for difference was 178% (95% CI 1235, 3379), 205% (95% CI 1539, 3319) and 169% (95% CI 1128, 2987) on days 1, 3 and 6 of the 6 day treatment, respectively. CONCLUSIONS: The extent of drug interaction by itraconazole was not time-dependent.


Subject(s)
Antifungal Agents/administration & dosage , Histamine H1 Antagonists, Non-Sedating/pharmacokinetics , Itraconazole/administration & dosage , Terfenadine/analogs & derivatives , Adult , Antifungal Agents/pharmacology , Cross-Over Studies , Drug Interactions , Female , Humans , Itraconazole/pharmacology , Japan , Male , Terfenadine/pharmacokinetics
11.
Ther Drug Monit ; 28(3): 321-5, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16778714

ABSTRACT

The objective of this study was to identify a single time-point for plasma lansoprazole measurement that adequately reflects area under the plasma lansoprazole concentration-time curve (AUC) after administration of lansoprazole alone or together with coadministration with CYP mediators. A randomized double-blind placebo-controlled crossover study design in 3 phases was conducted at intervals of 2 weeks. Eighteen healthy Japanese volunteers, comprising 3 CYP2C19 genotype groups, took a single oral 60-mg dose of lansoprazole after three 6-day pretreatments, that is, clarithromycin 800 mg/d, fluvoxamine 50 mg/d, and placebo. Blood samplings (10 mL each) for determination of lansoprazole were taken up to 24 hours after the administration of lansoprazole. Correlation between plasma lansoprazole concentrations at various time points and AUC0-24 were analyzed. Although there were significant differences in the pharmacokinetic parameters of lansoprazole during clarithromycin and placebo among CYP2C19 genotypes, the differences were not found during fluvoxamine. The plasma concentrations 3, 4, 6, and 8 hours after administration (C3, C4, C6, and C8, respectively) were highly correlated with AUC0-24 in coadministration with placebo, clarithromycin, and fluvoxamine (r>0.8, P<0.001). In particular, C6 showed a correlation coefficient of 0.940, 0.992, and 0.953 in coadministration with placebo, clarithromycin, and fluvoxamine, respectively, and was the most appropriate for estimating AUC0-24. The present study demonstrates that AUC of lansoprazole can be estimated by using a single time-point at C6. This method of plasma concentration monitoring at one time-point might be more suitable for AUC estimation than reference to CYP2C19 genotypes, particularly in coadministration of CYP mediators.


Subject(s)
Anti-Ulcer Agents/blood , Anti-Ulcer Agents/pharmacokinetics , Omeprazole/analogs & derivatives , 2-Pyridinylmethylsulfinylbenzimidazoles , Adult , Area Under Curve , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Clarithromycin/pharmacology , Cross-Over Studies , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Double-Blind Method , Drug Synergism , Enzyme Inhibitors/pharmacology , Female , Fluvoxamine/pharmacology , Genotype , Humans , Lansoprazole , Male , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Omeprazole/blood , Omeprazole/pharmacokinetics
12.
Eur J Clin Pharmacol ; 62(8): 597-603, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16783561

ABSTRACT

OBJECTIVE: Rabeprazole as a proton pump inhibitor (PPI) is mainly reduced to rabeprazole thioether via a nonenzymatic pathway, with minor CYP2C19 and CYP3A4 involvement. The aim of this study was to compare possible effects of clarithromycin and verapamil as inhibitors of CYP3A4 on the pharmacokinetics of rabeprazole among CYP2C19 genotypes. METHODS: A three-way randomized, double-blind, placebo-controlled crossover study was performed. Nineteen volunteers, of whom six were homozygous extensive metabolizers (EMs), eight were heterozygous EMs, and five were poor metabolizers (PMs) for CYP2C19, received three 6-day courses of either daily 800 mg clarithromycin, 240 mg verapamil, or placebo in a randomized fashion, with a single oral dose of 20 mg rabeprazole on day 6 in all cases. Plasma concentrations of rabeprazole and rabeprazole thioether were monitored up to 24 h after the dosing. RESULTS: In the control phase, the AUC(0-infinity) values for rabeprazole and rabeprazole thioether were 1,005+/-366 and 412+/-149 ng.h/ml in homozygous EMs, 1,108+/-340 and 491+/-245 ng.h/ml in heterozygous EMs, and 2,697+/-364 and 2,116+/-373 ng.h/ml in PMs, respectively. There were significant differences (p<0.001) in the AUC(0-infinity) of rabeprazole and rabeprazole thioether among three different CYP2C19 genotypes. In the clarithromycin and verapamil phases, no significant differences were found in the pharmacokinetic parameters of rabeprazole compared with those in the control phase irrespective of CYP2C19 genotypes, whereas the AUC(0-infinity) of rabeprazole thioether was significantly increased 2.8-fold and 2.3-fold in homozygous EMs (p<0.01), 2.0-fold and 2.0-fold in heterozygous EMs (p<0.05), and 1.6-fold and 1.9-fold in PMs (p<0.05), respectively. In each genotype group for CYP2C19, there were no statistical differences in the percent increase in those pharmacokinetic parameters between the clarithromycin and verapamil pretreatment phases. CONCLUSION: The pharmacokinetic parameters of rabeprazole were not altered by clarithromycin or verapamil irrespective of the CYP2C19 genotypes. However, this result shows that both clarithromycin and verapamil significantly influence the disposition of rabeprazole by inhibiting the oxidation of the thioether, since the AUC(0-infinity) of rabeprazole thioether that has no effect on acid secretion increased. Therefore, the pharmacokinetic interactions between rabeprazole and CYP3A4 or P-glycoprotein inhibitors have limited clinical significance.


Subject(s)
2-Pyridinylmethylsulfinylbenzimidazoles/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/genetics , Clarithromycin/pharmacology , Mixed Function Oxygenases/genetics , Verapamil/pharmacology , 2-Pyridinylmethylsulfinylbenzimidazoles/blood , 2-Pyridinylmethylsulfinylbenzimidazoles/metabolism , Area Under Curve , Aryl Hydrocarbon Hydroxylases/drug effects , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Double-Blind Method , Enzyme Inhibitors/pharmacology , Female , Genotype , Half-Life , Humans , Male , Metabolic Clearance Rate , Middle Aged , Mixed Function Oxygenases/drug effects , Rabeprazole
13.
Br J Clin Pharmacol ; 61(5): 533-7, 2006 May.
Article in English | MEDLINE | ID: mdl-16669846

ABSTRACT

AIM: To examine the effect of grapefruit juice, an inhibitor of CYP3A4 in the small intestine, on the disposition of manidipine enantiomers in healthy subjects. METHODS: A randomized cross-over study with at least a 2-week wash-out period was performed. Seven healthy male volunteers received an oral 40-mg dose of racemic manidipine after an overnight fast with either grapefruit juice (GFJ) or water, as a control study. Plasma concentrations of (S)- and (R)-manidipine were monitored up to 10 h after the dosing. RESULTS: The plasma concentrations of (S)-manidipine were significantly higher (P<0.001) than those of (R)-manidipine in the control phase with an S/R ratio for the AUC0-infinity of 1.62 (95% confidence interval 1.52, 1.73). GFJ significantly increased Cmax and AUC0-infinity of (S)-manidipine by 2.4-fold (P<0.01) and 2.3-fold (P<0.01), respectively, and Cmax and AUC0-infinity of (R)-manidipine were increased by 3.4-fold (P<0.01) and 3.0-fold (P<0.01), respectively. There were significant differences (P<0.01) in GFJ-mediated percentage increases in Cmax and AUC0-infinity of (S)-manidipine compared with those of (R)-manidipine. The S/R ratio for AUC0-infinity was significantly decreased from 1.6 to 1.2 during the GFJ phase (P<0.01). CONCLUSION: These results indicate that the stereoselective disposition of manidipine was altered by GFJ, as an inhibitor of CYP3A4. GFJ appears to affect this metabolic disposal of (R)-manidipine to a greater extent than that of (S)-manidipine.


Subject(s)
Beverages , Calcium Channel Blockers/pharmacokinetics , Citrus paradisi , Dihydropyridines/pharmacokinetics , Food-Drug Interactions , Administration, Oral , Adult , Area Under Curve , Cross-Over Studies , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Dihydropyridines/blood , Humans , Male , Nitrobenzenes , Piperazines , Stereoisomerism , Time Factors
14.
Br J Clin Pharmacol ; 61(5): 538-44, 2006 May.
Article in English | MEDLINE | ID: mdl-16669847

ABSTRACT

AIMS: Fexofenadine is a substrate of several drug transporters including P-glycoprotein. Our objective was to evaluate the possible effects of two P-glycoprotein inhibitors, itraconazole and diltiazem, on the pharmacokinetics of fexofenadine, a putative probe of P-glycoprotein activity in vivo, and compare the inhibitory effect between the two in healthy volunteers. METHODS: In a randomized three-phase crossover study, eight healthy volunteers were given oral doses of 100 mg itraconazole twice daily, 100 mg diltiazem twice daily or a placebo capsule twice daily (control) for 5 days. On the morning of day 5 each subject was given 120 mg fexofenadine, and plasma concentrations and urinary excretion of fexofenadine were measured up to 48 h after dosing. RESULTS: Itraconazole pretreatment significantly increased mean (+/-SD) peak plasma concentration (Cmax) of fexofenadine from 699 (+/-366) ng ml-1 to 1346 (+/-561) ng ml-1 (95% CI of differences 253, 1040; P<0.005) and the area under the plasma concentration-time curve [AUC0,infinity] from 4133 (+/-1776) ng ml-1 h to 11287 (+/-4552) ng ml-1 h (95% CI 3731, 10575; P<0.0001). Elimination half-life and renal clearance in the itraconazole phase were not altered significantly compared with those in the control phase. In contrast, diltiazem pretreatment did not affect Cmax (704+/-316 ng ml-1, 95% CI -145, 155), AUC0, infinity (4433+/-1565 ng ml-1 h, 95% CI -1353, 754), or other pharmacokinetic parameters of fexofenadine. CONCLUSIONS: Although some drug transporters other than P-glycoprotein are thought to play an important role in fexofenadine pharmacokinetics, itraconazole pretreatment increased fexofenadine exposure, probably due to the reduced first-pass effect by inhibiting the P-glycoprotein activity. As diltiazem pretreatment did not alter fexofenadine pharmacokinetics, therapeutic doses of diltiazem are unlikely to affect the P-glycoprotein activity in vivo.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Diltiazem/pharmacology , Histamine H1 Antagonists/pharmacokinetics , Itraconazole/pharmacology , Terfenadine/analogs & derivatives , Adult , Analysis of Variance , Cross-Over Studies , Drug Administration Schedule , Drug Interactions , Female , Histamine H1 Antagonists/blood , Histamine H1 Antagonists/urine , Humans , Male , Terfenadine/blood , Terfenadine/pharmacokinetics , Terfenadine/urine , Time Factors
15.
Article in English | MEDLINE | ID: mdl-16497565

ABSTRACT

A simple and sensitive column-switching high-performance liquid chromatographic method was developed for the simultaneous determination of omeprazole and its two main metabolites, 5-hydroxyomeprazole and omeprazole sulfone, in human plasma. Omeprazole, its two metabolites and lansoprazol as an internal standard were extracted from 1 ml of alkalinized plasma sample using diethyl ether-dichloromethane (45:55, v/v). The extract was injected into a column I (TSK-PW precolumn, 10 microm, 35 mm x 4.6 mm i.d.) for clean-up and column II (Inertsil ODS-80A column, 5 microm, 150 mm x 4.6mm i.d.) for separation. The mobile phase consisted of phosphate buffer-acetonitrile (92:8 v/v, pH 7.0) for clean-up and phosphate buffer-acetonitrile-methanol (65:30:5 v/v/v, pH 6.5) for separation, respectively. The peak was detected with an ultraviolet detector set at a wavelength of 302 nm, and total time for chromatographic separation was approximately 25 min. The validated concentration ranges of this method were 3-2000 ng/ml for omeprazole, 3-50 ng/ml for 5-hydroxyomeprazole and 3-1000 ng/ml for omeprazole sulfone. Mean recoveries were 84.3% for omeprazole, 64.3% for 5-hydroxyomeprazole and 86.1% for omeprazole sulfone. Intra- and inter-day coefficient variations were less than 5.1 and 6.6% for omeprazole, 4.6 and 5.0% for 5-hydroxyomeprazole and 4.6 and 4.9% for omeprazole sulfone at the different concentrations. The limits of quantification were 3 ng/ml for omeprazole and its metabolites. This method was suitable for use in pharmacokinetic studies in human volunteers, and provides a useful tool for measuring CYP2C19 activity.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Chromatography, High Pressure Liquid/methods , Mixed Function Oxygenases/genetics , Omeprazole/blood , Cytochrome P-450 CYP2C19 , Female , Genotype , Humans , Hydrogen-Ion Concentration , Male , Omeprazole/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Spectrophotometry, Ultraviolet
16.
Br J Clin Pharmacol ; 61(3): 309-14, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16487224

ABSTRACT

AIMS: Rabeprazole is known to be a substrate of CYP2C19. Our objective was to evaluate the possible effect of an inhibitor of CYP2C19, fluvoxamine, and compare the inhibitory effect of fluvoxamine on the metabolism of rabeprazole between CYP2C19 genotypes. METHODS: A two-way randomized double-blind, placebo-controlled crossover study was performed. Twenty-one volunteers, of whom seven were homozygous extensive metabolizers (EMs), eight were heterozygous EMs and six were poor metabolizers (PMs) for CYP2C19, received two 6-day courses of either fluvoxamine 50 mg or placebo daily in a randomized fashion with a single oral dose of rabeprazole 20 mg on day 6 in all cases. Plasma concentrations of rabeprazole and its metabolite rabeprazole thioether were monitored up to 24 h after dosing. RESULTS: During placebo administration, the mean AUCs(0,infinity) of rabeprazole in homozygous EMs, heterozygous EMs and PMs were 882 (95% CI, 602, 1162) ng ml-1h , 1214 (975, 1453) ng ml-1 h and 2762 (2482, 3042) ng ml-1 h (P<0.001), respectively. Fluvoxamine treatment increased AUC(0,infinity) of rabeprazole and rabeprazole thioether by 2.8-fold (P<0.001) and 5.1-fold (P<0.01) in homozygous EMs, and by 1.7-fold (P<0.01) and 2.6-fold (P<0.01) in heterozygous EMs, and significantly prolonged the elimination half-life of rabeprazole and rabeprazole thioether in homozygous EMs and in heterozygous EMs, whereas no difference in any pharmacokinetic parameters was found in PMs. There was a significant difference in fluvoxamine-mediated percentage increase in AUC(0,infinity) of rabeprazole and rabeprazole thioether between CYP2C19 genotypes. CONCLUSIONS: The present study indicates that there are significant drug interactions between rabeprazole and fluvoxamine in EMs of CYP2C19. It is predominantly involved in rabeprazole and rabeprazole thioether metabolism in EMs. Therefore, CYP2C19 is the key determinant of rabeprazole disposition in EMs.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Benzimidazoles/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Fluvoxamine/pharmacokinetics , Mixed Function Oxygenases/genetics , Omeprazole/analogs & derivatives , 2-Pyridinylmethylsulfinylbenzimidazoles , Administration, Oral , Adult , Area Under Curve , Benzimidazoles/blood , Benzimidazoles/metabolism , Cross-Over Studies , Cytochrome P-450 CYP2C19 , Double-Blind Method , Enzyme Inhibitors/blood , Enzyme Inhibitors/metabolism , Female , Fluvoxamine/administration & dosage , Fluvoxamine/adverse effects , Heterozygote , Homozygote , Humans , Male , Omeprazole/blood , Omeprazole/metabolism , Omeprazole/pharmacokinetics , Rabeprazole , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/adverse effects , Selective Serotonin Reuptake Inhibitors/pharmacokinetics
17.
J Chromatogr B Analyt Technol Biomed Life Sci ; 824(1-2): 238-43, 2005 Sep 25.
Article in English | MEDLINE | ID: mdl-16087412

ABSTRACT

A new sensitive column-switching high-performance liquid chromatographic (HPLC) method with ultraviolet detection was developed for the simultaneous determination of rabeprazole, a proton pump inhibitor, and its active metabolite, rabeprazole thioether in human plasma. Rabeprazole, its thioether metabolite and 5-methyl-2-[(4-(3-methoxypropoxy)-3-methyl pyridin-2-yl) methyl sulfinyl]-1H-benzimidazole, as an internal standard were extracted from 1 ml of plasma using diethyl ether-dichloromethane (9:1, v/v) mixture and the extract was injected into a column I (TSK-PW precolumn, 10 microm, 35 mmx4.6mm I.D.) for clean-up and column II (C18 Grand ODS-80TM TS analytical column, 5 microm, 250 mmx4.6 mm I.D.) for separation. The peak was detected with an ultraviolet detector set at a wavelength of 288 nm, and the total time for chromatographic separation was approximately 25 min. Mean absolute recoveries were 78.0 and 88.3% for rabeprazole and rabeprazole thioether, respectively. Intra- and inter-day coefficient variations were less than 6.5 and 4.5% for rabeprazole, 3.6 and 5.3% for rabeprazole thioether, respectively, at the different concentration ranges. The validated concentration ranges of this method were 1-1000 ng/ml for rabeprazole and 3-500 ng/ml for rabeprazole thioether. The limits of quantification were 1 ng/ml for rabeprazole and 3 ng/ml for rabeprazole thioether. The method was suitable for therapeutic drug monitoring and was applied to pharmacokinetic study in human volunteers.


Subject(s)
Benzimidazoles/blood , Chromatography, High Pressure Liquid/methods , Omeprazole/analogs & derivatives , Sulfides/blood , 2-Pyridinylmethylsulfinylbenzimidazoles , Administration, Oral , Adult , Area Under Curve , Benzimidazoles/metabolism , Benzimidazoles/pharmacokinetics , Female , Half-Life , Humans , Male , Omeprazole/blood , Omeprazole/metabolism , Omeprazole/pharmacokinetics , Rabeprazole , Reproducibility of Results , Sulfides/chemistry
18.
Br J Clin Pharmacol ; 60(1): 61-8, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15963095

ABSTRACT

AIMS: Lansoprazole is affected by polymorphism of CYP2C19. The aim of this study was to examine the effects of fluvoxamine, a CYP2C19 inhibitor, on the pharmacokinetics of each lansoprazole enantiomer among three different CYP2C19 genotype groups. METHODS: Eighteen healthy subjects, of whom six each were homozygous extensive metabolizers (homEMs), heterozygous extensive metabolizers (hetEMs), or poor metabolizers (PMs) for CYP2C19, participated in the study. Each subject received either placebo or fluvoxamine, 25 mg twice daily for 6 days, then a single oral dose of 60 mg of racemic lansoprazole. The plasma concentrations of lansoprazole enantiomers and lansoprazole sulphone were subsequently measured for 24 h post lansoprazole administration using liquid chromatography. RESULTS: In the homEMs and hetEMs, fluvoxamine significantly increased the AUC(0, infinity) and C(max) and prolonged the elimination half-life of both (R)- and (S)-lansoprazole, whereas in the PMs, the only statistically significant effect of fluvoxamine was on the AUC(0, infinity) for (R)-lansoprazole. The mean fluvoxamine-mediated percent increase in the AUC(0, infinity) of (R)-lansoprazole in the homEMs compared with the PMs was significant (P = 0.0117); however, C(max) did not differ among the three CYP2C19 genotypes. On the other hand, fluvoxamine induced a significant percent increase in both the AUC(0, infinity) and C(max) for (S)-lansoprazole in the homEMs compared with the hetEMs (P = 0.0007 and P = 0.0125, respectively) as well as compared with the PMs (P < 0.0001 for each parameter). The mean R : S ratio for AUC(0, infinity) of lansoprazole in the homEMs was significantly different between the placebo and the fluvoxamine treatment groups (12.7 (9.1, 16.8) vs 6.4 (5.4, 7.4), respectively, P < 0.0001), though not in the PMs (5.5 (4.3, 6.7) vs 5.9 (5.3, 6.5), respectively). CONCLUSIONS: The magnitude of the contribution of CYP2C19 to the metabolism of (S)-lansoprazole is much greater compared with that of the (R)-enantiomer. In extensive metabolizers, hepatic CYP2C19 plays an important role in the absorption and elimination of lansoprazole, particularly the (S)-enantiomer.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Enzyme Inhibitors/pharmacokinetics , Fluvoxamine/pharmacology , Mixed Function Oxygenases/genetics , Omeprazole/analogs & derivatives , 2-Pyridinylmethylsulfinylbenzimidazoles , Adult , Area Under Curve , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Cross-Over Studies , Cytochrome P-450 CYP2C19 , Dexlansoprazole , Double-Blind Method , Drug Interactions , Female , Genotype , Humans , Lansoprazole , Male , Mixed Function Oxygenases/antagonists & inhibitors , Omeprazole/pharmacokinetics
20.
Chirality ; 17(6): 338-44, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15856433

ABSTRACT

The aim of this study was to examine the effect of clarithromycin, a CYP3A4 inhibitor, on the enantioselective disposition of lansoprazole among three different CYP2C19 genotype groups in healthy Japanese subjects. These subjects included 6 each of homozygous extensive metabolizers (homEMs), heterozygous extensive metabolizers (hetEMs), and poor metabolizers (PMs). In the EMs of CYP2C19, clarithromycin markedly increased Cmax and the AUC0-infinity of (S)-lansoprazole and (S)-hydroxylansoprazole compared with those of the corresponding (R)-enantiomers. Clarithromycin significantly increased Cmax and the AUC0-infinity of (S)-lansoprazole in the homEMs by 110% and 115%, respectively, and in the hetEMs by 105% and 103%, respectively, compared with placebo. Furthermore, clarithromycin slightly prolonged the elimination half-life of (R)-lansoprazole in the homEMs and hetEMs but did not alter that of (S)-lansoprazole. In the of PMs CYP2C19, clarithromycin significantly increased Cmax and the AUC0-infinity and significantly prolonged the elimination half-lives of (R)- and (S)-lansoprazole by 51% and 49%, respectively. The present study suggests that there are significant drug interactions between (R)- or (S)-lansoprazole and clarithromycin in EMs by inhibiting the CYP3A4-catalyzed sulfoxidation primarily during the first pass, whereas in PMs, the overall metabolism of lansoprazole is inhibited.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Clarithromycin/pharmacology , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Omeprazole/analogs & derivatives , 2-Pyridinylmethylsulfinylbenzimidazoles , Adult , Clarithromycin/administration & dosage , Cross-Over Studies , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Dexlansoprazole , Double-Blind Method , Drug Interactions , Enzyme Inhibitors/pharmacology , Genotype , Heterozygote , Homozygote , Humans , Lansoprazole , Omeprazole/administration & dosage , Omeprazole/chemistry , Omeprazole/metabolism , Proton Pump Inhibitors , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...