Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 665
Filter
Add more filters

Publication year range
1.
Drug Metab Dispos ; 52(8): 911-918, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38849209

ABSTRACT

Arsenite is an important heavy metal. Some Chinese traditional medicines contain significant amounts of arsenite. The aim of this study was to investigate subacute exposure of arsenite on activities of cytochrome P450 enzymes and pharmacokinetic behaviors of drugs in rats. Midazolam, tolbutamide, metoprolol, omeprazole, caffeine, and chlorzoxazone, the probe substrates for cytochrome P450 (CYP) s3A, 2C6, 2D, 2C11, 1A, and 2E, were selected as probe drugs for the pharmacokinetic study. Significant decreases in areas under the curves of probe substrates were observed in rats after consecutive 30-day exposure to As at 12 mg/kg. Microsomal incubation study showed that the subacute exposure to arsenite resulted in little change in effects on the activities of P450 enzymes examined. However, everted gut sac study demonstrated that such exposure induced significant decreases in intestinal absorption of these drugs by both passive diffusion and carrier-mediated transport. In addition, in vivo study showed that the arsenite exposure decreased the rate of peristaltic propulsion. The decreases in intestinal permeability of the probe drugs and peristaltic propulsion rate most likely resulted in the observed decreases in the internal exposure of the probe drugs. Exposure to arsenite may lead to the reduction of the efficiencies of pharmaceutical agents coadministered resulting from the observed drug-drug interactions. SIGNIFICANCE STATEMENT: Exposure to arsenite may lead to the reduction of the efficiencies of pharmaceutical agents coadministered resulting from the observed drug-drug interactions. The present study, we found that P450 enzyme probe drug exposure was reduced in arsenic-exposed animals (areas under the curve) and the intestinal absorption of the drug was reduced in the animals. Subacute arsenic exposure tends to cause damage to intestinal function, which leads to reduced drug absorption.


Subject(s)
Arsenites , Cytochrome P-450 Enzyme System , Drug Interactions , Rats, Sprague-Dawley , Animals , Arsenites/toxicity , Arsenites/pharmacokinetics , Male , Rats , Cytochrome P-450 Enzyme System/metabolism , Intestinal Absorption/drug effects , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Omeprazole/pharmacology , Omeprazole/pharmacokinetics , Midazolam/pharmacokinetics , Caffeine/pharmacokinetics , Chlorzoxazone/pharmacokinetics , Metoprolol/pharmacokinetics , Metoprolol/pharmacology , Tolbutamide/pharmacokinetics , Sodium Compounds/toxicity , Sodium Compounds/pharmacokinetics
2.
Biol Pharm Bull ; 47(5): 1028-1032, 2024.
Article in English | MEDLINE | ID: mdl-38797695

ABSTRACT

Omeprazole, a gastric acid pump inhibitor, is repeatedly administered and is oxidatively metabolized mainly by polymorphic cytochrome P450 2C19. The prescribed dosage of omeprazole was discontinued or reduced in 47 of the 135 patients who received omeprazole alone in this survey, as recorded in the Japanese Adverse Drug Event Report database. The days to onset of omeprazole-related disorders were 3-4 d (median) and 16 d for intravenous 20-40 mg and oral 20 mg daily doses, respectively, in 34 patients for whom relevant data were available. The maximum plasma concentration of omeprazole was pharmacokinetically modeled after a single oral 40-mg dose in P450 2C19-defective poor metabolizers and was 2.4-fold higher than that in extensive metabolizers. The modeled area under the hepatic concentration curves of omeprazole in P450 2C19 poor metabolizers after virtual daily 40-mg doses for 7 d was 5.2-fold higher than that in the extensive metabolizers. Omeprazole-induced P450 2C19 (approx. 2-fold), resulting in increased hepatic intrinsic clearance in repeated doses, was considered after the second day. Virtual plasma/hepatic exposure estimated using pharmacokinetic modeling in subjects with P450 2C19 poor metabolizers indicated that these exposure levels virtually estimated could be one of causal factors for unexpected hepatic disorders induced by prescribed omeprazole, such as those resulting from drug interactions with repeatedly co-administered medicines.


Subject(s)
Cytochrome P-450 CYP2C19 , Liver , Omeprazole , Proton Pump Inhibitors , Humans , Adverse Drug Reaction Reporting Systems , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/blood , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C19/metabolism , Databases, Factual , East Asian People , Japan , Liver/metabolism , Liver/drug effects , Models, Biological , Omeprazole/pharmacokinetics , Omeprazole/adverse effects , Omeprazole/blood , Omeprazole/administration & dosage , Proton Pump Inhibitors/adverse effects , Proton Pump Inhibitors/administration & dosage , Proton Pump Inhibitors/pharmacokinetics , Proton Pump Inhibitors/blood
3.
Endocr Pract ; 30(6): 513-520, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38554774

ABSTRACT

OBJECTIVE: This study evaluates the impact of a representative proton pump inhibitor (PPI) (omeprazole), administered simultaneously or staggered, on the pharmacokinetics of levothyroxine (LT4) solution (Tirosint-SOL). METHODS: This was a randomized, 3-way crossover, comparative bioavailability study in 36 healthy adults under fasting conditions. Omeprazole 40 mg delayed-release capsule was administered once daily from Day 1 to 6 (mornings, Treatment-A; evenings, Treatment-B; none, Treatment-C) to increase and stabilize gastric pH. In the morning of Day 5, a single dose of LT4 solution 600 mcg was administered. Blood samples were collected 0 to 48 hours post-LT4 administration. Noncompartmental pharmacokinetic parameters were calculated for total serum thyroxine using baseline-corrected data. Maximum concentration (Cmax) and area under the concentration-time curve (AUC0-48) were included in an analysis of variance to obtain geometric mean ratios and 90% confidence intervals. RESULTS: For both comparisons (A/C and B/C), geometric mean ratios and 90% confidence intervals for all parameters were within the equivalence boundaries (80%-125%), indicating bioequivalence: for A/C, AUC0-48 98.98% [94%-104%], and Cmax 91.68% [87%-97%]; for B/C, AUC0-48 98.94% [95%-103%], and Cmax 94.90% [90%-100%]. Median Tmax (time associated with Cmax) was similar across treatments. CONCLUSION: This study demonstrated that Tirosint-SOL bioavailability is unaffected by coadministration of a representative PPI, given simultaneously or staggered by about 12 hours, compared to administration of LT4 solution alone. For hypothyroid patients on PPI therapy, administration of LT4 solution may reduce variations in thyroid stimulating hormone levels related to intermittent use of acid-reducing drugs and consequently the need for dose adjustments.


Subject(s)
Biological Availability , Cross-Over Studies , Omeprazole , Proton Pump Inhibitors , Thyroxine , Adult , Female , Humans , Male , Middle Aged , Young Adult , Drug Interactions , Omeprazole/pharmacokinetics , Omeprazole/administration & dosage , Proton Pump Inhibitors/administration & dosage , Proton Pump Inhibitors/pharmacokinetics , Proton Pump Inhibitors/pharmacology , Thyroxine/pharmacokinetics , Thyroxine/administration & dosage , Thyroxine/blood
4.
J Clin Psychopharmacol ; 43(2): 113-121, 2023.
Article in English | MEDLINE | ID: mdl-36700734

ABSTRACT

PURPOSE/BACKGROUND: Glycine transporter-1 inhibitors may ameliorate cognitive deficits in schizophrenia. This study evaluated potential drug-drug interactions with the glycine transporter-1 inhibitor BI 425809. METHODS/PROCEDURES: Interactions with cytochromes P450 (CYP) and P-glycoprotein (P-gp) were assessed in in vitro assays using human hepatocytes and Caco-2 cells, respectively. Pharmacokinetic characteristics of probe drugs were subsequently assessed in a Phase I, open-label, single-sequence crossover study in healthy male participants. Participants received a probe-drug cocktail containing midazolam (CYP3A4), warfarin (CYP2C9), and omeprazole (CYP2C19) and a separate dose of digoxin (P-gp), alone and on a background of steady-state BI 425809 25 mg once daily in 2 treatment periods. Adverse events were monitored. FINDINGS/RESULTS: In vitro assays revealed concentration-dependent induction of CYP3A4 and inhibition of P-gp by BI 425809. In the clinical study, 12 of 13 participants completed both periods. With BI 425809, area under the plasma concentration curve from administration to the last measurement (AUC 0-tz ) and maximum plasma concentration ( Cmax ) for midazolam were lower than when administered alone. Adjusted geometric mean ratios (90% confidence interval) were 70.6% (63.9%-78.1%) for AUC 0-tz and 77.6% (67.3%-89.4%) for Cmax . For warfarin and digoxin, AUC 0-tz and Cmax were similar with and without BI 425809. For omeprazole, BI 425809 slightly reduced AUC 0-tz but not Cmax versus omeprazole alone. No new safety signals were identified. IMPLICATIONS/CONCLUSIONS: These findings indicate induction of CYP3A4 by once-daily BI 425809 25 mg (the assumed highest therapeutic dose) and no meaningful effects on CYP2C9, CYP2C19, or P-gp in vivo.


Subject(s)
Glycine Plasma Membrane Transport Proteins , Midazolam , Humans , Male , Cytochrome P-450 CYP2C19 , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Cytochrome P-450 CYP3A , Warfarin , Cross-Over Studies , Cytochrome P-450 CYP2C9 , Caco-2 Cells , Caffeine/pharmacokinetics , Drug Interactions , Cytochrome P-450 Enzyme System/metabolism , Omeprazole/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B , Digoxin/pharmacokinetics , Area Under Curve
5.
Br J Clin Pharmacol ; 89(12): 3659-3668, 2023 12.
Article in English | MEDLINE | ID: mdl-37464978

ABSTRACT

AIMS: This study evaluated the effects of SHR0302 on the pharmacokinetics of cytochrome P450 (CYP) probe substrates. METHODS: We performed a single-centre, open-label, three-period drug-drug interaction (DDI) study in 24 healthy subjects (NCT05392127). Subjects received a single oral dose of 5 mg warfarin (CYP2C9), 20 mg omeprazole (CYP2C19) and 15 mg midazolam (CYP3A4) on Days 1, 8 and 22, and received 0.5 mg repaglinide (CYP2C8) on Days 7, 14 and 28. Multiple oral doses of 8 mg SHR0302 were administered once daily from Day 8 to Day 28. RESULTS: The exposure of S-warfarin and repaglinide were comparable before and after SHR0302 administration. AUC of midazolam was not affected by SHR0302, whereas the administration of SHR0302 slightly decreased the Cmax of midazolam by 7.6% (single dose) and 15.7% (once daily for 14 days). The AUC0-t , AUC0-inf , and Cmax of omeprazole were slightly decreased after a single dose of SHR0302 by 19.2%, 21.8% and 23.5%, respectively. In the presence of SHR0302 for 14 days, the AUC0-t , AUC0-inf , and Cmax of omeprazole were marginally reduced by 3.0%, 16.4% and 8.3%, respectively. According to the induction mechanism of the CYP enzyme, for the investigation of the induction effect, the results of multiple administrations of the perpetrator were more reliable than those of the single dose. CONCLUSIONS: The results demonstrated that co-administration of SHR0302 8 mg once daily is unlikely to have a clinically meaningful effect on the exposure of drugs metabolized by CYP3A4, CYP2C8, CYP2C9 and CYP2C19 in healthy subjects.


Subject(s)
Cytochrome P-450 CYP3A , Midazolam , Humans , Cytochrome P-450 CYP3A/metabolism , Midazolam/pharmacokinetics , Cytochrome P-450 CYP2C8/metabolism , Cytochrome P-450 CYP2C9 , Warfarin , Cytochrome P-450 CYP2C19/genetics , Drug Interactions , Cytochrome P-450 Enzyme System/metabolism , Omeprazole/pharmacokinetics , Healthy Volunteers
6.
Pharm Res ; 40(7): 1735-1750, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37226024

ABSTRACT

PURPOSE: To develop a whole physiologically based pharmacokinetic-pharmacodynamic (PBPK-PD) model to describe the pharmacokinetics and anti-gastric acid secretion of omeprazole in CYP2C19 extensive metabolizers (EMs), intermediate metabolizers (IMs), poor metabolizers (PMs) and ultrarapid metabolizers (UMs) following oral or intravenous administration. METHODS: A PBPK/PD model was built using Phoenix WinNolin software. Omeprazole was mainly metabolized by CYP2C19 and CYP3A4 and the CYP2C19 polymorphism was incorporated using in vitro data. We described the PD by using a turn-over model with parameter estimates from dogs and the effect of a meal on the acid secretion was also implemented. The model predictions were compared to 53 sets of clinical data. RESULTS: Predictions of omeprazole plasma concentration (72.2%) and 24 h stomach pH after administration (85%) were within 0.5-2.0-fold of the observed values, indicating that the PBPK-PD model was successfully developed. Sensitivity analysis demonstrated that the contributions of the tested factors to the plasma concentration of omeprazole were Vmax,2C19 ≈ Papp > Vmax,3A4 > Kti, and contributions to its pharmacodynamic were Vmax,2C19 > kome > kms > Papp > Vmax,3A4. The simulations showed that while the initial omeprazole dose in UMs, EMs, and IMs increased 7.5-, 3- and 1.25-fold compared to those of PMs, the therapeutic effect was similar. CONCLUSIONS: The successful establishment of this PBPK-PD model highlights that pharmacokinetic and pharmacodynamic profiles of drugs can be predicted using preclinical data. The PBPK-PD model also provided a feasible alternative to empirical guidance for the recommended doses of omeprazole.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Omeprazole , Humans , Animals , Dogs , Omeprazole/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 CYP2C19/genetics , Polymorphism, Genetic , Pharmaceutical Preparations , Genotype
7.
Mol Pharm ; 18(4): 1792-1805, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33739838

ABSTRACT

Human liver microsomes (HLM) and human hepatocytes (HH) are important in vitro systems for studies of intrinsic drug clearance (CLint) in the liver. However, the CLint values are often in disagreement for these two systems. Here, we investigated these differences in a side-by-side comparison of drug metabolism in HLM and HH prepared from 15 matched donors. Protein expression and intracellular unbound drug concentration (Kpuu) effects on the CLint were investigated for five prototypical probe substrates (bupropion-CYP2B6, diclofenac-CYP2C9, omeprazole-CYP2C19, bufuralol-CYP2D6, and midazolam-CYP3A4). The samples were donor-matched to compensate for inter-individual variability but still showed systematic differences in CLint. Global proteomics analysis outlined differences in HLM from HH and homogenates of human liver (HL), indicating variable enrichment of ER-localized cytochrome P450 (CYP) enzymes in the HLM preparation. This suggests that the HLM may not equally and accurately capture metabolic capacity for all CYPs. Scaling CLint with CYP amounts and Kpuu could only partly explain the discordance in absolute values of CLint for the five substrates. Nevertheless, scaling with CYP amounts improved the agreement in rank order for the majority of the substrates. Other factors, such as contribution of additional enzymes and variability in the proportions of active and inactive CYP enzymes in HLM and HH, may have to be considered to avoid the use of empirical scaling factors for prediction of drug metabolism.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Drug Evaluation, Preclinical/methods , Hepatocytes/enzymology , Liver/enzymology , Microsomes, Liver/enzymology , Bupropion/pharmacokinetics , Cytochrome P-450 Enzyme System/analysis , Diclofenac/pharmacokinetics , Ethanolamines/pharmacokinetics , Hepatobiliary Elimination , Humans , Liver/cytology , Midazolam/pharmacokinetics , Omeprazole/pharmacokinetics , Proteome/analysis , Proteomics
8.
Br J Clin Pharmacol ; 87(12): 4608-4618, 2021 12.
Article in English | MEDLINE | ID: mdl-33890704

ABSTRACT

We investigated the effect of deglucuronidation on the plasma concentration of the constituents of the Basel phenotyping cocktail and on the interpretation of the phenotyping results under basal conditions and after cytochrome P450 (CYP) induction with metamizole. The cocktail containing caffeine (CYP1A2), efavirenz (CYP2B6), flurbiprofen (CYP2C9), omeprazole (CYP2C19), metoprolol (CYP2D6) and midazolam (CYP3A4) was administered to 12 healthy subjects before (basal) and after treatment with metamizole for 1 week. In the basal state, deglucuronidation caused an increase in the plasma concentrations and area under the curve (AUC) of metoprolol, 8'-hydroxyefavirenz, 4'-hydroxyflurbiprofen and 1'-hydroxymidazolam. This effect could be visualized in Bland-Altman plots, where the values for 8'-hydroxyefavirenz, 4'-hydroxyflurbiprofen and 1'-hydroxymidazolam were mostly above the +20% threshold. As a result, the metabolic ratio (MR), calculated as AUCparent drug /AUCmetabolite , decreased with deglucuronidation for CYP2B6, CYP2C9 and CYP3A4 and increased for CYP2D6. Treatment with metamizole, a constitutive androstane receptor-dependent inducer of CYP2B6, CYP2C9, CYP2C19 and CYP3A4, accentuated the effect of deglucuronidation on AUC and MR. The correlation of MRs calculated as the plasma concentration ratio parent drug/metabolite with the MR calculated as the AUC ratio showed that 1 sample obtained between 2 and 6 hours after cocktail ingestion and analysed with and without deglucuronidation is sufficient to obtain reliable phenotyping results. Importantly, CYP2C9 and 3A4 induction would have been missed without deglucuronidation of the plasma samples. In conclusion, deglucuronidation of the plasma samples improves the stability of the phenotyping results of the Basel phenotyping cocktail and is necessary to reliably detect CYP induction.


Subject(s)
Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System , Glucuronides , Caffeine , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme System/metabolism , Drug Combinations , Flurbiprofen/pharmacokinetics , Glucuronides/metabolism , Humans , Metoprolol/pharmacokinetics , Midazolam/pharmacokinetics , Omeprazole/pharmacokinetics
9.
Pharm Res ; 38(3): 415-428, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33686560

ABSTRACT

PURPOSE: For decades, inflammation has been considered a cause of pharmacokinetic variability, mainly in relation to the inhibitory effect of pro-inflammatory cytokines on the expression level and activity of cytochrome P450 (CYP). In vitro and clinical studies have shown that two major CYPs, CYP2C19 and CYP3A4, are both impaired. The objective of the present study was to quantify the impact of the inflammatory response on the activity of both CYPs in order to predict the pharmacokinetic profile of their substrates according to systemic C-reactive protein (CRP). METHODS: The relationships between CRP concentration and both CYPs activities were estimated and validated using clinical data first on midazolam then on voriconazole. Finally, clinical data on omeprazole were used to validate the findings. For each substrate, a physiologically based pharmacokinetics model was built using a bottom-up approach, and the relationships between CRP level and CYP activities were estimated by a top-down approach. After incorporating the respective relationships, we compared the predictions and observed drug concentrations. RESULTS: Changes in pharmacokinetic profiles and parameters induced by inflammation seem to be captured accurately by the models. CONCLUSIONS: These findings suggest that the pharmacokinetics of CYP2C19 and CYP3A4 substrates can be predicted depending on the CRP concentration.


Subject(s)
Antifungal Agents/pharmacokinetics , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP3A/metabolism , Inflammation/drug therapy , Computer Simulation , Cytochrome P-450 Enzyme Inhibitors/pharmacokinetics , Drug Interactions , Humans , Midazolam/pharmacokinetics , Models, Biological , Omeprazole/pharmacokinetics , Voriconazole/pharmacokinetics
10.
Biol Pharm Bull ; 44(6): 762-770, 2021.
Article in English | MEDLINE | ID: mdl-34078808

ABSTRACT

A cocktail study is an in vivo evaluation method to assess multiple CYP activities via a single trial and single administration of a cocktail drug that is a combination of multiple CYP substrates. However, multiple blood samples are required to evaluate the pharmacokinetics of a CYP probe drug. A limited-point sampling method is generally beneficial in clinical studies because of the simplified protocol and reduced participant burden. The aim of this study was to evaluate whether a limited-point plasma concentration analysis of CYP substrates in a cocktail drug could predict their area under the curve (AUC). We created prediction models of five CYP substrates (caffeine, losartan, omeprazole, dextromethorphan, and midazolam) using multiple linear regressions from the data of two cocktail studies, and then performed predictability analysis of these models using data derived from data in the co-administration with inducer (rifampicin) and inhibitors (fluvoxamine and cimetidine). For the administration of inhibitors, the AUC prediction accuracy (mean absolute error (MAE)) were <39.5% in Model 1 and <26.2% in Model 2 which were created using 1- and 4-point sampling data. MAE shows larger values in the administration of inducer in compared with the administration of inhibitors. The accuracy of the prediction in Model 2 could be acceptable for screening of inhibitions. MAE for caffeine, dextromethorphan, and midazolam were acceptable in the model that used 4 sampling points from all data. The use of this method could reduce the burden on the subject and make it possible to evaluate each AUC in a minimally invasive manner.


Subject(s)
Area Under Curve , Cytochrome P-450 Enzyme System/metabolism , Models, Biological , Administration, Oral , Adult , Caffeine/blood , Caffeine/pharmacokinetics , Dextromethorphan/blood , Dextromethorphan/pharmacokinetics , Humans , Losartan/blood , Losartan/pharmacokinetics , Male , Midazolam/blood , Midazolam/pharmacokinetics , Omeprazole/blood , Omeprazole/pharmacokinetics , Young Adult
11.
AAPS PharmSciTech ; 22(5): 188, 2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34159427

ABSTRACT

Omeprazole is a widely used over-the-counter (20 mg) proton pump inhibitor, usually supplied as oral enteric-coated pellets intended to release at pH 5.5 and higher; however, it is sensitive to acidic pH. The likelihood of elevated gastric pH in practice is very high for patients; thus, the aim of this study was to investigate the effect of elevated pH on the performance of commercial omeprazole pellets. Commercial enteric-coated delayed-release pellets were tested with water uptake-weight loss (WU-WL) test at pH range between 1.2 and 4.5 in addition to "gastric" (pH 1.2 or 4.5) and "intestinal" (pH 7.4) phase dissolution tests. The range of physical characteristics of pellets was determined with a single pellet size and sedimentation time measurement, followed by the application of modified Stokes' Law equation. The coefficient of variation of pellet size and density, and volume-density determination coefficient (R2) as descriptors of coating thickness and microstructure variability, degree of ionisation of enteric polymers, aqueous solubility and molecular weight of plasticisers have been found useful to explain commercial delayed-release pellets behaviour during WU-WL and dissolution test. Investigated commercial delayed-release pellets demonstrated pH-dependent WU-WL results. "Gastric phase" dissolution testing of pellets at pH 4.5 showed the highest omeprazole degradation (48.1%) for Nosch Labs, intermediate values of dose loss (23.4% and 17.1%) for Teva and UQUIFA delayed-release pellets, respectively. Lab Liconsa pellets have been found as the least susceptible (3.2% of dose loss). Additionally, "gastric phase" dissolution test at pH 4.5 significantly influenced omeprazole release during the "intestinal phase". The risk of inadequate therapy associated with intake of investigated enteric-coated delayed-release pellets at elevated gastric pH has been found as minimal for Lab Liconsa and has increased from UQUIFA and Teva to Nosh Labs pellets.


Subject(s)
Drugs, Generic/chemistry , Gastrointestinal Absorption/drug effects , Omeprazole/chemistry , Patents as Topic , Proton Pump Inhibitors/chemistry , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Drugs, Generic/pharmacokinetics , Gastrointestinal Absorption/physiology , Humans , Hydrogen-Ion Concentration , Male , Omeprazole/pharmacokinetics , Proton Pump Inhibitors/pharmacokinetics , Solubility , Tablets, Enteric-Coated , Young Adult
12.
Am J Ther ; 26(3): e308-e313, 2019.
Article in English | MEDLINE | ID: mdl-28452843

ABSTRACT

BACKGROUND: Proton pump inhibition (PPI) administrated together with dabigatran reduces the risk of gastrointestinal hemorrhage. However, there is a discussion regarding possible PPI-dabigatran interaction that may reduce the efficacy of this therapy. STUDY QUESTION: To determine the impact of concomitant PPI on dabigatran plasma levels in patients with nonvalvular atrial fibrillation (NV-AF). STUDY DESIGN: A pilot prospective study in patients with NV-AF on dabigatran therapy was performed; 31 patients were enrolled. PPI with either omeprazole or pantoprazole was administrated in 19 patients. MEASURES AND OUTCOMES: Blood samples were taken for the assessment of the dabigatran trough and peak levels. Dabigatran concentration was measured with the Hemoclot Thrombin Inhibitor Assay. RESULTS: There were significant differences in dabigatran trough level comparing patients treated with PPI and patients without PPI (58.86 ± 36.76 ng/mL vs. 110.72 ± 88.47 ng/mL, P < 0.05). Similarly, there were significant differences in dabigatran peak level between compared groups (88.0 ± 20.5 ng/mL vs. 174.4 ± 139.64 ng/mL, P < 0.05). CONCLUSIONS: This pilot study demonstrated the interaction between PPI and dabigatran levels in patients with NV-AF.


Subject(s)
Anticoagulants/pharmacokinetics , Atrial Fibrillation/drug therapy , Dabigatran/pharmacokinetics , Gastrointestinal Hemorrhage/prevention & control , Proton Pump Inhibitors/pharmacokinetics , Aged , Aged, 80 and over , Anticoagulants/adverse effects , Dabigatran/adverse effects , Drug Interactions , Female , Gastrointestinal Hemorrhage/chemically induced , Humans , Male , Middle Aged , Omeprazole/administration & dosage , Omeprazole/pharmacokinetics , Pantoprazole/administration & dosage , Pantoprazole/pharmacokinetics , Pilot Projects , Prospective Studies , Proton Pump Inhibitors/administration & dosage
13.
J Pharmacol Sci ; 139(4): 361-366, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30902567

ABSTRACT

Polymorphisms of cytochrome P450 (CYP) enzymes can affect enzymatic activity, drug metabolism and drug interactions. Although the potential for drug interactions is especially important when co-administering drugs with strong inductive or inhibitory potential towards drug-metabolizing enzymes, the relationship between CYP genotypes and the extent of the inductive or inhibitory effects remain poorly understood. We investigated the effects of rifampicin (inductive) and fluvoxamine (inhibitory) on metabolism of omeprazole and CYP2C19 enzymatic activity in 19 healthy Japanese subjects. Pharmacokinetic analyses of the CYP2C19 probe drug, omeprazole, were performed before and after rifampicin or fluvoxamine administration. The allele frequencies of the CYP2C19*1, CYP2C19*2 and CYP2C19*3 genotypes were 65.8%, 26.3% and 7.9%, respectively. Subjects with the CYP2C19*1 allele displayed higher levels of omeprazole metabolism than those without the CYP2C19*1 allele. Rifampicin increased omeprazole metabolism in all subjects irrespective of genotype, which suggested that CYP2C19 enzymatic activity was induced by rifampicin administration for all genotypes. Conversely, while fluvoxamine reduced omeprazole metabolism in subjects carrying the CYP2C19*1 allele, it had no impact on omeprazole pharmacokinetics in subjects without this allele. The genotyping of CYP2C19 may be useful for predicting drug interactions with metabolic inhibitors.


Subject(s)
Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C19/metabolism , Fluvoxamine/pharmacology , Genetic Association Studies , Omeprazole/pharmacokinetics , Proton Pump Inhibitors/pharmacokinetics , Rifampin/pharmacology , Adult , Alleles , Drug Interactions , Fluvoxamine/administration & dosage , Genotype , Humans , Male , Omeprazole/administration & dosage , Proton Pump Inhibitors/administration & dosage , Rifampin/administration & dosage , Young Adult
14.
Support Care Cancer ; 27(3): 819-827, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30084103

ABSTRACT

PURPOSE: Rolapitant is a neurokinin-1 receptor antagonist indicated in combination with other antiemetic agents in adults for the prevention of delayed chemotherapy-induced nausea and vomiting. We evaluated the effects of rolapitant oral on the pharmacokinetics of probe substrates for cytochrome P450 (CYP) 2D6 (dextromethorphan), 2C9 (tolbutamide), 2C19 (omeprazole), 2B6 (efavirenz), and 2C8 (repaglinide) in healthy subjects. METHODS: This open-label, multipart, randomized, phase 1 study assessed cohorts of 20-26 healthy subjects administered dextromethorphan, tolbutamide plus omeprazole, efavirenz, or repaglinide with and without single, oral doses of rolapitant. Maximum plasma analyte concentrations (Cmax) and area under the plasma analyte concentration-time curves (AUC) were estimated using noncompartmental analysis, and geometric mean ratios (GMRs) and 90% confidence intervals for the ratios of test (rolapitant plus probe substrate) to reference (probe substrate alone) treatment were calculated. RESULTS: Rolapitant significantly increased the systemic exposure of dextromethorphan in terms of Cmax and AUC0-inf by 2.2- to 3.3-fold as observed in GMRs on days 7 and 14. Rolapitant did not affect systemic exposure of tolbutamide, and minor excursions outside of the 80-125% no effect limits were detected for omeprazole, efavirenz, and repaglinide. CONCLUSIONS: Inhibition of dextromethorphan by a single oral dose of rolapitant 180 mg is clinically significant and can last at least 7 days. No clinically significant interaction was observed between rolapitant and substrates of CYP2C9, CYP2C19, CYP2B6, or CYP2C8. CYP2D6 substrate drugs with a narrow therapeutic index may require monitoring for adverse reactions if given concomitantly with rolapitant.


Subject(s)
Antiemetics/pharmacology , Neurokinin-1 Receptor Antagonists/pharmacology , Spiro Compounds/pharmacology , Administration, Oral , Adolescent , Adult , Alkynes , Benzoxazines/pharmacokinetics , Carbamates/pharmacokinetics , Cyclopropanes , Cytochrome P-450 CYP2B6/drug effects , Cytochrome P-450 CYP2C19/drug effects , Cytochrome P-450 CYP2C8/drug effects , Cytochrome P-450 CYP2C9/drug effects , Cytochrome P-450 CYP2D6/drug effects , Dextromethorphan/pharmacokinetics , Drug Combinations , Drug Interactions , Female , Healthy Volunteers , Humans , Male , Middle Aged , Molecular Probes/pharmacokinetics , Neurokinin-1 Receptor Antagonists/administration & dosage , Omeprazole/pharmacokinetics , Piperidines/pharmacokinetics , Tolbutamide/pharmacokinetics , Young Adult
15.
Xenobiotica ; 49(8): 975-980, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30215539

ABSTRACT

1. In clinical practice, diammonium glycyrrhizinate is usually used with omeprazole in patients with viral hepatitis and cirrhosis accompanied by peptic ulcers. However, the drug interaction between diammonium glycyrrhizinate and omeprazole remains unclear. 2. In this study, the effects of diammonium glycyrrhizinate on the pharmacokinetics of omeprazole was investigated by liquid chromatography-tandem mass spectrometry (LC-MS/MS) analytical method. Male Sprague-Dawley rats were randomly assigned to two groups: omeprazole and omeprazole + diammonium glycyrrhizinate, and the main pharmacokinetic parameters were estimated after oral administration. It was found that using the omeprazole along with the diammonium glycyrrhizinate increased the AUC, AUMC, Cmax for omeprazole. 3. For this reason, we used the LC-MS/MS to detect the binding rate of plasma protein (BRPP) of omeprazole in rats, it was found that diammonium glycyrrhizinate could decrease the BRPP in rats. In addition, we found that diammonium glycyrrhizinate specifically inhibited the enzyme activity of the CYP2C19 and CYP3A4, which are involved in the metabolism of the omeprazole. 4. These results mean that diammonium glycyrrhizinate could inhibit the metabolism and increase the plasma concentration of the omeprazole in rats. Overall, diammonium glycyrrhizinate can influence the pharmacokinetics of omeprazole by inhibiting CYP2C19 and CYP3A4 activities and decreasing BRPP of omeprazole.


Subject(s)
Blood Proteins/metabolism , Cytochrome P-450 Enzyme System/metabolism , Glycyrrhizic Acid/pharmacology , Omeprazole/pharmacokinetics , Animals , Feasibility Studies , Male , Protein Binding/drug effects , Rats, Sprague-Dawley , Reference Standards
16.
Biopharm Drug Dispos ; 40(3-4): 135-150, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30921829

ABSTRACT

Physiologically based pharmacokinetic (PBPK) modelling and simulation is a useful tool in predicting the PK profiles of a drug, assessing the effects of covariates such as demographics, ethnicity, genetic polymorphisms and disease status on the PK, and evaluating the potential of drug-drug interactions. We developed a Korean-specific virtual population for the SimCYP® Simulator (version 15 used) and evaluated the population's predictive performance using six substrate drugs (midazolam, S-warfarin, metoprolol, omeprazole, lorazepam and rosuvastatin) of five major drug metabolizing enzymes (DMEs) and two transporters. Forty-three parameters including the proportion of phenotypes in DMEs and transporters were incorporated into the Korean-specific virtual population. The simulated concentration-time profiles in Koreans were overlapped with most of the observed concentrations for the selected substrate drugs with a < 2-fold difference in clearance. Furthermore, we found some drug models within the SimCYP® library can be improved, e.g., the minor allele frequency of ABCG2 and the fraction metabolized by UGT2B15 should be incorporated for rosuvastatin and lorazepam, respectively. The Korean-specific population can be used to evaluate the impact of ethnicity on the PKs of a drug, particularly in various stages of drug development.


Subject(s)
Asian People , Models, Biological , Software , Adult , Computer Simulation , Cytochrome P-450 Enzyme System/metabolism , Female , Glucuronosyltransferase/metabolism , Humans , Liver-Specific Organic Anion Transporter 1/metabolism , Lorazepam/pharmacokinetics , Male , Metoprolol/pharmacokinetics , Midazolam/pharmacokinetics , Middle Aged , Omeprazole/pharmacokinetics , Rosuvastatin Calcium/pharmacokinetics , Warfarin/pharmacokinetics , Young Adult
17.
Molecules ; 24(7)2019 Mar 27.
Article in English | MEDLINE | ID: mdl-30934659

ABSTRACT

In this study, a novel sort of sample preparation sorbent was developed, by preparing thin layer graphene oxide tablets (GO-Tabs) utilizing a mixture of graphene oxide and polyethylene glycol on a polyethylene substrate. The GO-Tabs were used for extraction and concentration of omeprazole (OME) in human saliva samples. The determination of OME was carried out using liquid chromatography-tandem mass spectrometry (LC⁻MS/MS) under gradient LC conditions and in the positive ion mode (ESI+) with mass transitions of m/z 346.3→198.0 for OME and m/z 369.98→252.0 for the internal standard. Standard calibration for the saliva samples was in the range of 2.0⁻2000 nmol L-1. Limits of detection and quantification were 0.05 and 2.0 nmol L-1, respectively. Method validation showed good method accuracy and precision; the inter-day precision values ranged from 5.7 to 8.3 (%RSD), and the accuracy of determinations varied from -11.8% to 13.3% (% deviation from nominal values). The extraction recovery was 60%, and GO-Tabs could be re-used for more than ten extractions without deterioration in recovery. In this study, the determination of OME in real human saliva samples using GO-Tab extraction was validated.


Subject(s)
Graphite/chemistry , Omeprazole/pharmacokinetics , Oxides/chemistry , Pharmaceutical Preparations/chemistry , Saliva , Tablets/chemistry , Chromatography, Liquid , Electron Microscope Tomography , Humans , Omeprazole/chemistry , Pharmaceutical Preparations/isolation & purification , Polymerization , Reproducibility of Results , Solvents , Tandem Mass Spectrometry
18.
Pharm Biol ; 57(1): 449-452, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31290355

ABSTRACT

Context: Omeprazole and astragaloside IV (AS-IV) are widely used for the treatment of gastric ulcers in China clinics. Objective: This study investigates the effects of AS-IV on the pharmacokinetics of omeprazole in rats. Materials and methods: The pharmacokinetics of orally administered omeprazole (2 mg/kg), with or without AS-IV (100 mg/kg/day for 7 days) pretreatment, were investigated in male Sprague-Dawley rats (two groups of six animals each) using LC-MS/MS. A Caco-2 cell transwell model and rat liver microsome incubation systems were also used to support the in vivo pharmacokinetic data and investigate its potential mechanism. Results: The results indicated that co-administration of AS-IV could decrease the systemic exposure of omeprazole significantly (p < 0.05), including AUC0-t (717.20 ± 177.63 vs. 1166.25 ± 186.65 ng h/mL) and Cmax (272.35 ± 25.81 vs. 366.34 ± 32.57 ng/mL). The t1/2 of omeprazole also decreased significantly (1.78 ± 0.15 vs. 2.23 ± 0.27 h, p < 0.05). The efflux ratio of omeprazole across the Caco-2 cell transwell model increased significantly from 1.73 to 2.67 (p < 0.05), and the metabolic stability of omeprazole was decreased from 42.6 ± 7.8 to 26.2 ± 5.1 min with the pretreatment of AS-IV (p < 0.05). Discussion and conclusions: AS-IV could decrease the systemic exposure of omeprazole in rats when AS-IV and omeprazole were co-administered, and it might exert these effects through decreasing the absorption of omeprazole by inducing P-gp, or through accelerating the metabolism of omeprazole in rat liver by inducing the activity of CYP3A4.


Subject(s)
Omeprazole/pharmacokinetics , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Caco-2 Cells , Humans , Male , Microsomes, Liver/drug effects , Rats , Rats, Sprague-Dawley
19.
Prague Med Rep ; 120(1): 5-9, 2019.
Article in English | MEDLINE | ID: mdl-31103048

ABSTRACT

Data on gastric pH in rats to be used in preclinical models for pH-dependent drug absorption are still limited or contradictory. The aim of this study was to describe gastric pH in rats at fasted state and to evaluate its changes induced by pentagastrin or omeprazole in order to mimic gastric pH at fasted and fed human subjects. Twenty Wistar rats, fasting for 12 h, were randomly assigned into four treatment groups (n=5): control, pre-treated with omeprazole 2 h before pH measurement, pre-treated with omeprazole 12 h before pH measurement, and pre-treated with pentagastrin 20 min before pH measurement. An incision on the stomach wall was made in anesthetized animals, and pH of gastric juice was measured. The observed pH values were significantly different among groups (p=0.0341), with the median (IQR) values of gastric pH of 3.5 (2.7-4.2), 6.7 (4.7-7.0), 5.6 (3.5-6.4) and 2.2 (1.6-3.1) in control, omeprazole 2 h, omeprazole 12 h and pentagastin group, respectively. We recommend using short interval pentagastrin and 2 h omeprazole pre-treatment in fasting animals to model similar gastric pH as is expected in human fasted and fed state pharmacokinetic studies, respectively.


Subject(s)
Gastric Mucosa , Omeprazole , Proton Pump Inhibitors , Stomach , Animals , Humans , Hydrogen-Ion Concentration , Models, Animal , Omeprazole/pharmacokinetics , Proton Pump Inhibitors/pharmacokinetics , Random Allocation , Rats , Rats, Wistar , Stomach/chemistry
20.
Br J Clin Pharmacol ; 84(5): 997-1005, 2018 05.
Article in English | MEDLINE | ID: mdl-29377228

ABSTRACT

AIMS: Although substantial progress has been made in understanding of ontogeny of drug metabolism, there is still a gap of knowledge in developmental pharmacogenetics in neonates. We hypothesized that both age and pharmacogenetics might explain the developmental pattern of CYP2C19. We conducted a population pharmacokinetic-pharmacogenetic study to quantify the developmental pharmacogenetics of CYP2C19 in neonates and young infants using omeprazole as a probe drug. METHODS: Pharmacokinetic samples were collected from 51 Caucasian neonates and young infants, who were receiving omeprazole treatment. Population pharmacokinetic-pharmacogenetic analysis of omeprazole and its metabolites was performed using NONMEM. RESULTS: Data fitted a one-compartment parent and metabolite model with first-order absorption and elimination. CYP2C19 and CYP3A4 are predominantly involved in the metabolism of omeprazole despite their relatively low activities compared to adults. The clearance of omeprazole converted to 5-hydroxy-omeprazole (CLOMZ-M1 ) increases with postnatal age. In CYP2C19 poor and intermediate metabolizers, model-predicted CLOMZ-M1 are 12.5% (5-95% percentile: 3-14.9%) and 44.9% (5-95% percentile: 29.9-72.6%) of the value in extensive/ultrarapid metabolizer, respectively. Model-predicted absorption rate constant of omeprazole is 6.93 (5-95% percentile: 3.01-14.61) times higher in ABCB1 homozygous mutant patients, 1.86 (5-95% percentile: 0.86-3.47) times higher in ABCB1 heterozygous patients than that in ABCB1 homozygous wild-type patients. CONCLUSIONS: Developmental pharmacogenetics of CYP2C19 was quantitatively described in neonates and young infants using omeprazole as a probe drug. Our findings emphasize the importance of semiphysiological developmental pharmacokinetic modelling approach when evaluating developmental pharmacogenetics of drugs with multiple routes of biotransformation.


Subject(s)
Cytochrome P-450 CYP2C19/genetics , Omeprazole/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Age Factors , Female , Genotype , Heterozygote , Homozygote , Humans , Infant , Infant, Newborn , Male , Models, Biological , Omeprazole/blood , Phenotype , Polymorphism, Genetic , Proton Pump Inhibitors/blood , Proton Pump Inhibitors/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL