Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 281
Filter
Add more filters

Publication year range
1.
Planta Med ; 90(10): 792-800, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39013429

ABSTRACT

This work investigated interactions ascribed to the administration of phytomedicines containing Valeriana officinalis and Piper methysticum with conventional drugs. The phytomedicines were characterized by HPLC and administered per os to male Wistar rats, either concomitantly or not with the CYP3A substrate midazolam. To distinguish between the presystemic or systemic effect, midazolam was given orally and intravenously. The effects on the P-gp substrate fexofenadine uptake by Caco-2 cells were examined. The valerenic acid content was 1.6 ± 0.1 mg per tablet, whereas kavain was 13.7 ± 0.3 mg/capsule. Valerian and kava-kava extracts increased the maximum plasma concentration (Cmax) of midazolam 2- and 4-fold compared to the control, respectively. The area under the plasma concentrations versus time curve (AUC(0-∞)) was enhanced from 994.3 ± 152.3 ng.h/mL (control) to 3041 ± 398 ng.h/mL (valerian) and 4139 ± 373 ng.h/mL (kava-kava). The half-life of midazolam was not affected. These changes were attributed to the inhibition of midazolam metabolism by the enteric CYP3A since the i. v. pharmacokinetic of midazolam remained unchanged. The kava-kava extract augmented the uptake of fexofenadine by 3.5-fold compared to the control. Although Valeriana increased the uptake of fexofenadine, it was not statistically significant to that of the control (12.5 ± 3.7 ng/mg protein vs. 5.4 ± 0.3 ng/mg protein, respectively). Therefore, phytomedicines containing V. officinalis or P. methysticum inhibited the intestinal metabolism of midazolam in rats. Conversely, the P-gp-mediated transport of fexofenadine was preferably affected by kava-kava.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1 , Cytochrome P-450 CYP3A , Kava , Midazolam , Plant Extracts , Rats, Wistar , Terfenadine , Valerian , Animals , Valerian/chemistry , Midazolam/pharmacokinetics , Midazolam/pharmacology , Male , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A/drug effects , Plant Extracts/pharmacology , Plant Extracts/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Terfenadine/analogs & derivatives , Terfenadine/pharmacokinetics , Humans , Caco-2 Cells , Rats , Kava/chemistry , Herb-Drug Interactions , Piper/chemistry , Indenes , Pyrones , Sesquiterpenes
2.
Ther Drug Monit ; 45(4): 539-545, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36645711

ABSTRACT

BACKGROUND: Fexofenadine is a recommended in vivo probe drug for phenotyping P-glycoprotein (P-gp) and organic anion transporting polypeptide (OATP) 1B1/3 transporter activities. This study evaluated a limited sampling strategy using a population pharmacokinetic approach to estimate plasma fexofenadine exposure as an index of P-gp and OATP activities. METHODS: In a previous study, a single oral dose of fexofenadine (120 mg) was administered alone or in combination with grapefruit juice, Panax ginseng , or Echinacea purpurea to healthy adult participants. Serial plasma samples were collected up to 72 hours after administration and fexofenadine concentrations were measured. A population pharmacokinetic model was developed using nonlinear mixed-effects modeling. Limited sampling models (LSMs) using single and 2-timepoint fexofenadine concentrations were compared with full profiles from intense sampling using empirical Bayesian post hoc estimations of systemic exposure derived from the population pharmacokinetic model. Predefined criteria for LSM selection and validation included a coefficient of determination (R 2 ) ≥ 0.90, relative percent mean prediction error ≥ -5 to ≤5%, relative percent mean absolute error ≤ 10%, and relative percent root mean square error ≤ 15%. RESULTS: Fexofenadine concentrations (n = 1520) were well described using a 2-compartment model. Grapefruit juice decreased the relative oral bioavailability of fexofenadine by 25%, whereas P. ginseng and E. purpurea had no effect. All the evaluated single timepoint fexofenadine LSMs showed unacceptable percent mean prediction error, percent mean absolute error, and/or percent root mean square error. Although adding a second time point improved precision, the predefined criteria were not met. CONCLUSIONS: Identifying novel fexofenadine LSMs to estimate P-gp and OATP1B1/3 activities in healthy adults for future transporter-mediated drug-drug interaction studies remains elusive.


Subject(s)
Citrus paradisi , Organic Anion Transporters , Adult , Humans , Bayes Theorem , Terfenadine/pharmacokinetics , Pharmaceutical Preparations
3.
Pharm Res ; 38(4): 647-655, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33825113

ABSTRACT

PURPOSE: Fexofenadine is a well-identified in vivo probe substrate of P-glycoprotein (P-gp) and/or organic anion transporting polypeptide (OATP). This work aimed to investigate the transplacental pharmacokinetics of fexofenadine enantiomers with and without the selective P-gp inhibitor fluoxetine. METHODS: The chiral transplacental pharmacokinetics of fexofenadine-fluoxetine interaction was determined using the ex vivo human placenta perfusion model (n = 4). In the Control period, racemic fexofenadine (75 ng of each enantiomer/ml) was added in the maternal circuit. In the Interaction period, racemic fluoxetine (50 ng of each enantiomer/mL) and racemic fexofenadine (75 ng of each enantiomer/mL) were added to the maternal circulation. In both periods, maternal and fetal perfusate samples were taken over 90 min. RESULTS: The (S)-(-)- and (R)-(+)-fexofenadine fetal-to-maternal ratio values in Control and Interaction periods were similar (~0.18). The placental transfer rates were similar between (S)-(-)- and (R)-(+)-fexofenadine in both Control (0.0024 vs 0.0019 min-1) and Interaction (0.0019 vs 0.0021 min-1) periods. In both Control and Interaction periods, the enantiomeric fexofenadine ratios [R-(+)/S-(-)] were approximately 1. CONCLUSIONS: Our study showed a low extent, slow rate of non-enantioselective placental transfer of fexofenadine enantiomers, indicating a limited fetal fexofenadine exposure mediated by placental P-gp and/or OATP2B1. The fluoxetine interaction did not affect the non-enantioselective transplacental transfer of fexofenadine. The ex vivo placental perfusion model accurately predicts in vivo placental transfer of fexofenadine enantiomers with remarkably similar values (~0.17), and thus estimates the limited fetal exposure.


Subject(s)
Histamine H1 Antagonists, Non-Sedating/pharmacokinetics , Maternal-Fetal Exchange/drug effects , Placenta/metabolism , Terfenadine/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Area Under Curve , Drug Interactions , Female , Fluoxetine/administration & dosage , Fluoxetine/pharmacokinetics , Histamine H1 Antagonists, Non-Sedating/administration & dosage , Humans , Perfusion/instrumentation , Perfusion/methods , Pregnancy , Pregnancy Complications/drug therapy , Pregnancy Complications/immunology , Stereoisomerism , Terfenadine/administration & dosage , Terfenadine/pharmacokinetics
4.
J Pharmacol Sci ; 147(1): 95-103, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34294379

ABSTRACT

Several false-positive results in the human ether-à-gogo-related gene test suggest that blockers of the rapid component of delayed rectifier K+ current (IKr) do not necessarily produce drug-induced arrhythmias. Specifically, the occurrence of early afterdepolarization (EAD) differs among IKr blockers, even if the prolonged action potential duration is in the same range. To predict EAD in drug-induced arrhythmias, we proposed a prediction method based on the mechanisms underlying the difference in frequency of EAD among nonselective IKr blockers. The mechanisms were elucidated by examining how different blockade kinetics of L-type Ca2+ current (ICaL) affect the frequency of EAD, using mathematical models of human ventricular myocytes. Addition of voltage-independent ICaL blockade resulted in the suppression of EAD. However, when voltage-dependent ICaL blockade kinetics of amiodarone, bepridil, and terfenadine were incorporated into ICaL in the model, bepridil and terfenadine induced EAD more than the voltage-independent ICaL blockade, while amiodarone suppressed EAD more effectively. Opposite effects were accounted for by the difference in ICaL blockade at negatively polarized potential. EAD occurrence was found to be associated with ICaL blockade measured at -20 mV. These results suggest that voltage dependence of ICaL blockade may be useful in predicting the different risks of nonselective IKr blockers.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Potassium/metabolism , Action Potentials , Amiodarone/adverse effects , Amiodarone/pharmacokinetics , Bepridil/adverse effects , Bepridil/pharmacokinetics , Calcium Channels, L-Type/metabolism , Computer Simulation , Heart Ventricles/drug effects , Humans , Membrane Potentials/drug effects , Models, Theoretical , Myocytes, Cardiac , Terfenadine/adverse effects , Terfenadine/pharmacokinetics
5.
Acta Pharmacol Sin ; 42(3): 470-481, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32555444

ABSTRACT

P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) are involved in intestinal barrier. Short-chain fatty acids (SCFAs) play important roles in maintaining intestinal barrier. In this study we explored how SCFAs affected the expression and function of intestinal P-gp and BCRP in rats. Rats received 150 mM acetate, propionate or butyrate in drinking water for 4 weeks. In SCFA-treated rats, the expression and function of intestinal P-gp were decreased, but those of intestinal BCRP were increased; intestinal p-p65 was also decreased, which was positively related to P-gp protein expression. Among the three SCFAs tested, butyrate exhibited the strongest induction or inhibitory effect, followed by propionate and acetate. Similar results were observed in mouse primary enterocytes and Caco-2 cells treated with acetate (5 mM), propionate (2 mM), or butyrate (1 mM). In Caco-2 cells, addition of butyrate, vorinostat, and valproate (two classic HDAC inhibitors), Bay117082 (selective inhibitor of NF-κB activation) or NF-κB p65 silencing significantly decreased the expression of P-gp and the level of phosphorylated p65 (p-p65). Furthermore, butyrate attenuated the expression of P-gp and p-p65 induced by TNF-α (NF-κB activator) and theophylline (HDAC activator). However, vorinostat, valproate, Bay117082, TNF-α or p65 silencing hardly affected BCRP protein expression. But GW9662 (selective PPARγ antagonist) or PPARγ silencing abolished BCRP induction by butyrate and troglitazone (PPARγ agonist). SCFAs-treated rats showed higher intestinal protein expression of PPARγ, which was positively related to BCRP protein expression. Butyrate increased plasma exposure of fexofenadine but decreased that of rosuvastatin following oral dose to rats. In conclusion, SCFAs exert opposite effects on the expression and function of intestinal P-gp and BCRP; butyrate downregulated P-gp expression and function possibly via inhibiting HDAC/NF-κB pathways; butyrate induced BCRP expression and function partly via PPARγ activation.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Acetates/pharmacology , Butyrates/pharmacology , Intestinal Mucosa/metabolism , Propionates/pharmacology , Animals , Caco-2 Cells , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , Mice, Inbred BALB C , NF-kappa B/metabolism , PPAR gamma/metabolism , Rats, Sprague-Dawley , Rosuvastatin Calcium/pharmacokinetics , Signal Transduction/drug effects , Terfenadine/analogs & derivatives , Terfenadine/pharmacokinetics
6.
Xenobiotica ; 51(3): 366-372, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33256506

ABSTRACT

Breviscapine (BRE) is usually used for long-term use in patients with cardiovascular diseases such as coronary heart disease, angina pectoris, and cerebral thrombosis. It is possible to combine it with P-glycoprotein (P-gp) substrates in clinic. At present, little is known about whether the simultaneous use of BRE affects the disposal of P-gp substrates. The aim of this study was to evaluate the effect of BRE on the pharmacokinetics of fexofenadine (FEX), a P-gp probe substrate and its associations with the MDR1 C3435T genetic polymorphism in healthy volunteers. In this randomised, open-label, placebo-controlled, two-phase crossover clinical study, drug interactions were evaluated in healthy volunteers. FEX was used as a phenotypic probe for P-gp. In each phase, 18 volunteers were given daily doses of 120 mg (40 mg, three times a day) of BRE tablet or a placebo for 14 days. On day 15, a single oral dose of 120 mg FEX hydrochloride was given orally. Blood samples were collected at predefined time intervals, and plasma levels of FEX were determined by ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). The pharmacokinetic parameters were calculated by non-compartmental method, and bioequivalence was evaluated. Results showed that BRE pretreatment did not significantly affect the pharmacokinetics of FEX. The peak maximum plasma concentration (C max) and the area under the plasma concentration-time curve from zero to infinity (AUCinf) mean value of FEX with BRE and placebo-treated groups were 699 ng/mL vs. 710 ng/mL and 2972.5 ng⋅h/mL vs. 3460.5 ng⋅h/mL, respectively. The geometric mean ratios (90% confidence intervals) for FEX C max and AUCinf were within the pre-specified range of 0.8-1.25, indicating that FEX in the two pretreatment phases were bioequivalent. Pharmacokinetic parameters of FEX showed no statistically significant difference between MDR1 C3435T CC, CT and TT genotype, revealing that BRE and MDR1 C3435T gene polymorphisms did not affect the pharmacokinetics of FEX in healthy volunteers.


Subject(s)
Flavonoids/pharmacology , Polymorphism, Genetic , Terfenadine/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B/genetics , Area Under Curve , Cross-Over Studies , Healthy Volunteers , Humans , Tandem Mass Spectrometry , Terfenadine/pharmacokinetics
7.
Biomed Chromatogr ; 35(7): e5093, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33634891

ABSTRACT

A rapid and specific UPLC-MS/MS method with a total run time of 3.5 min was developed for the determination of pravastatin, fexofenadine, rosuvastatin, and methotrexate in rat primary hepatocytes. After protein precipitation with 70% acetonitrile (containing 30% H2 O), these four analytes were separated under gradient conditions with a mobile phase consisting of 0.03% acetic acid (v/v) and methanol at a flow rate of 0.50 mL/min. The linearity, recovery, matrix effect, accuracy, precision, and stability of the method were well validated. We evaluated drug-drug interactions based on these four compounds in freshly suspended hepatocytes. The hepatic uptake of pravastatin, fexofenadine, rosuvastatin, and methotrexate at 4°C was significantly lower than that at 37°C, and the hepatocytes were saturable with increased substrate concentration and culture time, suggesting that the rat primary hepatocyte model was successfully established. Triptolide showed a significant inhibitory effect on the hepatic uptake of these four compounds. In conclusion, this method was successfully employed for the quantification of pravastatin, fexofenadine, rosuvastatin, and methotrexate and was used to verify the rat primary hepatocyte model for Oatp1, Oatp2, Oatp4, and Oat2 transporter studies. Then, we applied this model to explore the effect of triptolide on these four transporters.


Subject(s)
Hepatocytes/metabolism , Methotrexate , Pravastatin , Rosuvastatin Calcium , Terfenadine/analogs & derivatives , Animals , Cells, Cultured , Chromatography, High Pressure Liquid/methods , Diterpenes/analysis , Diterpenes/pharmacokinetics , Drug Interactions , Epoxy Compounds/analysis , Epoxy Compounds/pharmacokinetics , Linear Models , Male , Methotrexate/analysis , Methotrexate/pharmacokinetics , Phenanthrenes/analysis , Phenanthrenes/pharmacokinetics , Pravastatin/analysis , Pravastatin/pharmacokinetics , Rats, Wistar , Reproducibility of Results , Rosuvastatin Calcium/analysis , Rosuvastatin Calcium/pharmacokinetics , Sensitivity and Specificity , Tandem Mass Spectrometry/methods , Terfenadine/analysis , Terfenadine/pharmacokinetics
8.
Pharm Res ; 37(7): 131, 2020 Jun 17.
Article in English | MEDLINE | ID: mdl-32557079

ABSTRACT

BACKGROUND AND OBJECTIVE: Fluoxetine, antidepressant widely-used during pregnancy, is a selective inhibitor for P-glycoprotein (P-gp). Fexofenadine, an in vivo P-gp probe, is an antihistamine drug for seasonal allergic rhinitis and chronic urticaria treatment during pregnancy and it is available as a racemic mixture. This study evaluated the chiral discrimination of P-gp investigating the effect of fluoxetine on maternal-fetal pharmacokinetics of fexofenadine. METHODS: Healthy parturient women received either a single oral dose of 60 mg racemic fexofenadine (Control group; n = 8) or a single oral dose of 40 mg racemic fluoxetine 3 h before a single oral dose of 60 mg racemic fexofenadine (Interaction group; n = 8). Maternal blood and urine samples were collected up to 48 h after fexofenadine administration. At delivery, maternal-placental-fetal blood samples were collected. RESULTS: The maternal pharmacokinetics of fexofenadine was enantioselective (AUC0-∞R-(+)/S-(-) ~ 1.5) in both control and interaction groups. Fluoxetine increased AUC0-∞ (267.7 vs 376.1 ng.h/mL) and decreased oral total clearance (105.1 vs 74.4 L/h) only of S-(-)-fexofenadine, whereas the renal clearance were reduced for both enantiomers, suggesting that the intestinal P-gp-mediated transport of S-(-)-fexofenadine is influenced by fluoxetine to a greater extent that the R-(+)-fexofenadine. However, the transplacental transfer of fexofenadine is low (~16%), non-enantioselective and non-influenced by fluoxetine. CONCLUSIONS: A single oral dose of 40 mg fluoxetine inhibited the intestinal P-gp mediated transport of S-(-)-fexofenadine to a greater extent than R-(+)-fexofenadine in parturient women. However, the placental P-gp did not discriminate fexofenadine enantiomers and was not inhibited by fluoxetine.


Subject(s)
Antidepressive Agents, Second-Generation/administration & dosage , Fluoxetine/administration & dosage , Histamine H1 Antagonists, Non-Sedating/pharmacokinetics , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Parturition , Terfenadine/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adult , Antidepressive Agents, Second-Generation/adverse effects , Case-Control Studies , Drug Interactions , Female , Fetal Blood/metabolism , Fluoxetine/adverse effects , Histamine H1 Antagonists, Non-Sedating/administration & dosage , Histamine H1 Antagonists, Non-Sedating/blood , Humans , Intestinal Mucosa/metabolism , Maternal-Fetal Exchange , Placental Circulation , Pregnancy , Terfenadine/administration & dosage , Terfenadine/blood , Terfenadine/pharmacokinetics , Young Adult
9.
Drug Metab Dispos ; 47(8): 832-842, 2019 08.
Article in English | MEDLINE | ID: mdl-31123035

ABSTRACT

Organic anion transporting polypeptide 2B1 (OATP2B1) is a widely expressed membrane transporter with diverse substrate specificity. In vitro and clinical studies suggest a role for intestinal OATP2B1 in the oral absorption of medications. Moreover, OATP2B1 is highly expressed in hepatocytes where it is thought to promote liver drug clearance. However, until now, a shortcoming of studies implicating OATP2B1 in drug disposition has been a lack of in vivo models. Here, we report the development of a knockout (KO) mouse model with targeted, global disruption of the Slco2b1 gene to examine the disposition of two confirmed mOATP2B1 substrates, namely, fexofenadine and rosuvastatin. The plasma pharmacokinetics of intravenously administered fexofenadine was not different between KO and wild-type (WT) mice. However, after oral fexofenadine administration, KO mice had 70% and 41% lower maximal plasma concentration (C max) and area under the plasma concentration-time curve (AUC0-last) than WT mice, respectively. In WT mice, coadministration of fexofenadine with grapefruit juice (GFJ) or apple juice (AJ) was associated with reduced C max by 80% and 88%, respectively, while the AUC0-last values were lower by 35% and 70%, respectively. In KO mice, AJ coadministration reduced oral fexofenadine C max and AUC0-last values by 67% and 59%, respectively, while GFJ had no effects. Intravenous and oral rosuvastatin pharmacokinetics were similar among WT and KO mice. We conclude that intestinal OATP2B1 is a determinant of oral fexofenadine absorption, as well as a target for fruit juice interactions. OATP2B1 does not significantly influence rosuvastatin disposition in mice. SIGNIFICANCE STATEMENT: A novel mouse model with targeted disruption of the Slco2b1 gene revealed that OATP2B1 is a determinant of oral absorption but not systemic disposition of fexofenadine, as well as a target of fruit juice interactions. Rosuvastatin oral and intravenous pharmacokinetics were not dependent on OATP2B1. These findings support the utility of the Slco2b1 KO mouse model for defining mechanisms of drug disposition at the intersection of in vitro and clinical pharmacology.


Subject(s)
Intestinal Mucosa/metabolism , Organic Anion Transporters/metabolism , Rosuvastatin Calcium/pharmacokinetics , Terfenadine/analogs & derivatives , Administration, Intravenous , Administration, Oral , Animals , Area Under Curve , Food-Drug Interactions , Fruit and Vegetable Juices , HEK293 Cells , HeLa Cells , Humans , Intestinal Absorption , Male , Mice , Mice, Knockout , Organic Anion Transporters/genetics , Rosuvastatin Calcium/administration & dosage , Terfenadine/administration & dosage , Terfenadine/pharmacokinetics
10.
Xenobiotica ; 48(4): 342-347, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28436281

ABSTRACT

1. Roles of human cytochrome P450 (P450) 3A4 in oxidation of an antihistaminic drug terfenadine have been previously investigated in association with terfenadine-ketoconazole interaction. Several antihistamine drugs have been recently identified as substrates for multiple P450 enzymes. In this study, overall roles of P450 3A4, 2J2, and 4F12 enzymes in terfenadine t-butyl hydroxylation were investigated in small intestines and livers from humans, marmosets, and/or cynomolgus monkeys. 2. Human liver microsomes and liver and small intestine microsomes from marmosets and cynomolgus monkeys effectively mediated terfenadine t-butyl hydroxylation. Ketoconazole and N-hydroxy-N'-(4-butyl-2-methylphenyl)-formamidine (a P450 4A/F inhibitor) almost completely and moderately inhibited these activities, respectively, in human liver microsomes; however, these chemicals did not show substantially suppression in marmoset liver. Anti-human P450 3A and 4F antibodies showed the roughly supportive inhibitory effects. 3. Recombinant P450 3A4/90 and 4F12 showed high terfenadine t-butyl hydroxylation activities with substrate inhibition constants of 84-144 µM (under 26-76 µM of Km values), in similar manners to liver and intestine microsomes. 4. These results suggest that human and marmoset P450 3A4/90 and 4F12 in livers or small intestines played important roles in terfenadine t-butyl hydroxylation. Marmosets could be a model for humans during first pass extraction of terfenadine and related substrates.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 CYP3A/metabolism , Intestine, Small/enzymology , Liver/enzymology , Terfenadine/pharmacokinetics , Animals , Humans , Hydroxylation , Macaca fascicularis , Microsomes, Liver/enzymology
11.
J Vet Pharmacol Ther ; 41(6): 805-814, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30020547

ABSTRACT

Thirty-two Collies were used to determine the impact of ABCB1 genotype and phenotype on the plasma pharmacokinetics of fexofenadine's (Fex) R- and S-enantiomers after bolus Fex administration, as human P-gp exhibits stereoselectivity. Each Collie's ABCB1 genotype and ivermectin (IVM) sensitivity (phenotype) was determined prior to study enrolment. Wild-type (WT) Collies had lower plasma concentrations of the individual enantiomers as compared to heterozygous IVM nonsensitive (HNS), heterozygous IVM-sensitive (HS) and homozygous mutant (MUT) Collies. Based on pairwise statistical comparison, WT Collies had statistically significantly lower (AUC0-last ) and peak (Cmax ) values compared to HS, HNS and MUT Collies. Tmax was not influenced by genotype/phenotype. Inter-individual variability in PK metrics tended to be largest for WT Collies. Although the influence of genotype/phenotype on Fex PK occurred with the individual isomers, impairment of S-Fex absorption, particularly in the MUT dogs, exceeded that associated with R-Fex. Since Fex elimination occurs primarily via biliary excretion via a transporter other than P-glycoprotein, and based upon our understanding of Fex absorption kinetics, we attributed these differences primarily to the absorption portion of the profile. These differences are expressed in a stereo-specific manner. These results demonstrate the potential negative impact on estimates of drug effectiveness and toxicity, especially for P-gp substrates that do not exhibit Central Nervous System toxicities.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Anti-Allergic Agents/pharmacokinetics , Dogs/genetics , Genotype , Terfenadine/analogs & derivatives , Animals , Anti-Allergic Agents/blood , Area Under Curve , Dogs/metabolism , Female , Half-Life , Male , Terfenadine/blood , Terfenadine/pharmacokinetics
12.
Drug Metab Dispos ; 45(5): 569-575, 2017 05.
Article in English | MEDLINE | ID: mdl-28188296

ABSTRACT

The aim of this study was to determine the effects of garlic and ginkgo herbal extracts on the pharmacokinetics of the P-glycoprotein (P-gp)/organic anion-transporting polypeptides (Oatps) substrate fexofenadine. Male rats were dosed orally with garlic (120 mg/kg), ginkgo (17 mg/kg), St. John's wort (SJW; 1000 mg/kg; positive control), or Milli-Q water for 14 days. On day 15, rats either were administered fexofenadine (orally or i.v.), had their livers isolated and perfused with fexofenadine, or had their small intestines divided into four segments (SI-SIV) and analyzed for P-gp and Oatp1a5. In vivo, SJW increased the clearance of i.v. administered fexofenadine by 28%. Garlic increased the area under the curve0-∞ and maximum plasma concentration of orally administered fexofenadine by 47% and 85%, respectively. Ginkgo and SJW had no effect on the oral absorption of fexofenadine. In the perfused liver, garlic, ginkgo, and SJW increased the biliary clearance of fexofenadine with respect to perfusate by 71%, 121%, and 234%, respectively. SJW increased the biliary clearance relative to the liver concentration by 64%. The ratio of liver to perfusate concentrations significantly increased in all treated groups. The expression of Oatp1a5 in SI was increased by garlic (88%) and SJW (63%). There were no significant changes in the expression of P-gp. Induction of intestinal Oatp1a5 by garlic may explain the increased absorption of orally administered fexofenadine. Ginkgo had no effect on the expression of intestinal P-gp or Oatp1a5. A dual inductive effect by SJW on opposing intestinal epithelial transport by Oatp1a5 and P-gp remains a possibility.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Garlic/chemistry , Ginkgo biloba/chemistry , Hypericum/chemistry , Organic Anion Transporters, Sodium-Independent/metabolism , Plant Extracts/pharmacology , Terfenadine/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Administration, Oral , Animals , Drug Interactions , Injections, Intravenous , Intestinal Mucosa/metabolism , Liver/metabolism , Male , Organic Anion Transporters, Sodium-Independent/genetics , Perfusion , Plant Extracts/isolation & purification , Rats , Substrate Specificity , Terfenadine/administration & dosage , Terfenadine/blood , Terfenadine/pharmacokinetics , Tissue Distribution
13.
Eur J Clin Pharmacol ; 73(3): 343-349, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27981349

ABSTRACT

PURPOSE: Piperine (PIP) has been found to inhibit P-glycoprotein (P-gp) function in rats, suggesting that it may have the potential to modulate P-gp-mediated drug efflux in humans. The aim of this study was to evaluate the effect of PIP on the pharmacokinetics of fexofenadine (FEX), a P-gp substrate, in healthy volunteers. METHODS: An open-label, two-period, sequential study involving 12 healthy volunteers was conducted. A single oral dose of FEX 120 mg was given to volunteers during the control phase and after the treatment phase. A once-daily oral dose of PIP 20 mg was given to volunteers during the treatment phase (10 days). Blood samples were collected at predefined time intervals, and plasma samples containing FEX were analyzed by liquid chromatography-tandem mass spectrometry. RESULTS: Treatment with PIP significantly increased maximum plasma concentration of FEX [406.9 (control) vs. 767 ng/mL (treatment)] and area under the plasma concentration-time curve [3403.7 (control) vs. 5724.7 ng.h/mL (treatment)] when compared to the control phase. In contrast, PIP treatment significantly decreased apparent oral clearance of FEX [35.4 (control) vs. 20.7 L/h (treatment)] as compared to the control. There was no significant change observed in the half life and renal clearance of FEX between the treatment phase and control phase. CONCLUSIONS: The results suggest that altered pharmacokinetics and enhanced bioavailability of FEX might be attributed to PIP-mediated inhibition of P-gp drug efflux. Therefore, intake of PIP or dietary supplements containing PIP may potentially enhance the absorption or bioavailability of P-gp substrate drugs in addition to FEX.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Alkaloids/pharmacology , Benzodioxoles/pharmacology , Histamine H1 Antagonists, Non-Sedating/pharmacokinetics , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Terfenadine/analogs & derivatives , Adult , Area Under Curve , Biological Availability , Half-Life , Healthy Volunteers , Histamine H1 Antagonists, Non-Sedating/pharmacology , Humans , Male , Terfenadine/pharmacokinetics , Terfenadine/pharmacology
14.
Xenobiotica ; 47(3): 230-235, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27151911

ABSTRACT

1. Diosmin (DSN) has been found to possess P-glycoprotein (P-gp) inhibition activity in vitro and in vivo in rats, which may have potential to cause P-gp-mediated interactions in humans. The purpose of the present study was to investigate the effect of DSN on pharmacokinetics of fexofenadine (FEX) in healthy human volunteers. 2. An open-label, two-period, sequential study was conducted in 12 healthy male volunteers. A single dose of FEX 120 mg was administered to volunteers during control and treatment phases. A single dose of DSN 500 mg was administered to volunteers daily for period of 10 days. The blood and urine samples were collected at predetermined time intervals after FEX dosing and analyzed by LC-MS/MS. 3. Treatment with DSN significantly increased the peak maximum plasma concentration (Cmax) and area under the plasma concentration-time curve (AUC) of FEX by 49.2% (523.28 versus 780.63 ng/mL) and 64.4% (3459.48 versus 5687.74 ng h/mL), respectively as compared to control phase. On the other hand, apparent oral clearance (CL/F) of FEX was significantly decreased by 41.3% (37.03 versus 21.75 L/h) and there was no significant change was observed in Tmax, T1/2, and CLr (renal clearance) upon treatment with DSN when compared to control. 4. The results suggest that altered pharmacokinetics of FEX might be attributed to DSN-mediated inhibition of P-gp-mediated efflux in humans. Therefore, we conclude that intake of DSN or dietary supplements containing DSN may potentially increase the absorption or bioavailability of FEX, a P-gp substrate in humans.


Subject(s)
Diosmin/metabolism , Terfenadine/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Biological Availability , Drug Interactions , Healthy Volunteers , Humans , Male , Terfenadine/pharmacokinetics
15.
Drug Dev Ind Pharm ; 43(6): 932-938, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28102715

ABSTRACT

BACKGROUND AND OBJECTIVE: Capsaicin is the main pungent principle present in chili peppers has been found to possess P-glycoprotein (P-gp) inhibition activity in vitro, which may have the potential to modulate bioavailability of P-gp substrates. Therefore, purpose of this study was to evaluate the effect of capsaicin on intestinal absorption and bioavailability of fexofenadine, a P-gp substrate in rats. METHODS: The mechanistic evaluation was determined by non-everted sac and intestinal perfusion studies to explore the intestinal absorption of fexofenadine. These results were confirmed by an in vivo pharmacokinetic study of oral administered fexofenadine in rats. RESULTS: The intestinal transport and apparent permeability (Papp) of fexofenadine were increased significantly by 2.8 and 2.6 fold, respectively, in ileum of capsaicin treated rats when compared to control group. Similarly, absorption rate constant (Ka), fraction absorbed (Fab) and effective permeability (Peff) of fexofenadine were increased significantly by 2.8, 2.9 and 3.4 fold, respectively, in ileum of rats pretreated with capsaicin when compared to control group. In addition, maximum plasma concentration (Cmax) and area under the concentration-time curve (AUC) were increased significantly by 2.3 and 2.4 fold, respectively, in rats pretreated with capsaicin as compared to control group. Furthermore, obtained results in rats pretreated with capsaicin were comparable to verapamil (positive control) treated rats. CONCLUSIONS: Capsaicin pretreatment significantly enhanced the intestinal absorption and bioavailability of fexofenadine in rats likely by inhibition of P-gp mediated cellular efflux, suggesting that the combined use of capsaicin with P-gp substrates may require close monitoring for potential drug interactions.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Capsaicin/pharmacology , Histamine H1 Antagonists/pharmacokinetics , Terfenadine/analogs & derivatives , Adrenergic beta-Antagonists/pharmacokinetics , Animals , Area Under Curve , Biological Availability , Histamine H1 Antagonists/administration & dosage , Ileum/drug effects , Ileum/metabolism , In Vitro Techniques , Intestinal Absorption/drug effects , Male , Perfusion , Permeability/drug effects , Propranolol/pharmacokinetics , Rats , Rats, Wistar , Terfenadine/administration & dosage , Terfenadine/pharmacokinetics
16.
Br J Clin Pharmacol ; 82(6): 1580-1590, 2016 12.
Article in English | MEDLINE | ID: mdl-27495955

ABSTRACT

AIMS: We assessed the drug interaction profile of fermented red ginseng with respect to the activity of major cytochrome (CYP) P450 enzymes and of a drug transporter protein, P-glycoprotein (P-gp), in healthy volunteers. METHODS: This study was an open-label crossover study. The CYP probe cocktail drugs caffeine, losartan, dextromethorphan, omeprazole, midazolam and fexofenadine were administered before and after 2 weeks of fermented red ginseng administration. Plasma samples were collected, and tolerability was assessed. Pharmacokinetic parameters were calculated, and the 90% confidence intervals (CIs) of the geometric mean ratios of the parameters were determined from logarithmically transformed data. Values were compared between before and after fermented red ginseng administration using analysis of variance (anova). RESULTS: Fifteen healthy male subjects were evaluated, none of whom were genetically defined as a poor CYP2C9, CYP2C19 or CYP2D6 metabolizer based on genotyping. Before and after fermented red ginseng administration, the geometric least-square mean metabolic ratio (90% CI) was 0.901 (0.830-0.979) for caffeine (CYP1A2) to paraxanthine, 0.774 (0.720-0.831) for losartan (CYP2C9) to EXP3174, 1.052 (0.925-1.197) for omeprazole (CYP2C19) to 5-hydroxyomeprazole, 1.150 (0.860-1.538) for dextromethorphan (CYP2D6) to dextrorphan, and 0.816 (0.673-0.990) for midazolam (CYP3A4) to 1-hydroxymidazolam. The geometric mean ratio of the area under the curve of the last sampling time (AUClast ) for fexofenadine (P-gp) was 1.322 (1.112-1.571). CONCLUSION: No significantly different drug interactions were observed between fermented red ginseng and the CYP probe substrates following the two-week administration of concentrated fermented red ginseng. However, the inhibition of P-gp was significantly different between fermented red ginseng and the CYP probe substrates. The use of fermented red ginseng requires close attention due to the potential for increased systemic exposure when it is used in combination with P-gp substrate drugs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP3A/metabolism , Fermented Foods , Panax , Pharmaceutical Preparations/metabolism , Adult , Caffeine/administration & dosage , Caffeine/pharmacokinetics , Cross-Over Studies , Drug Interactions , Healthy Volunteers , Humans , Losartan/administration & dosage , Losartan/pharmacokinetics , Male , Midazolam/administration & dosage , Midazolam/pharmacokinetics , Middle Aged , Omeprazole/administration & dosage , Omeprazole/pharmacokinetics , Pharmaceutical Preparations/administration & dosage , Terfenadine/administration & dosage , Terfenadine/analogs & derivatives , Terfenadine/pharmacokinetics , Young Adult
17.
Eur J Clin Pharmacol ; 72(7): 797-805, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27023466

ABSTRACT

PURPOSE: In Caco-2 cells, folate uptake via the proton-coupled folate transporter (PCFT) increases significantly by a 3-day treatment with 1,25-dihydroxyvitamin D3 (1,25(OH)2D3). Additionally, mRNA content and protein expression of the transporter OATP1A2 were increased up to ninefold with 1,25(OH)2D3. We investigated whether these in vitro findings can be confirmed in humans in vivo. METHODS: Ten healthy volunteers (six women) received 5 mg folic acid orally once before and once together with the last intake of a 10-day course of 0.5 µg 1,25(OH)2D3 orally. One hundred twenty milligrams fexofenadine, an OATP1A2 substrate, was taken in 1 day before the first folic acid intake, and again on the ninth day of 1,25(OH)2D3 intake. Duodenal biopsies were taken for transporter mRNA assessments once before and once on the ninth or tenth day of the vitamin D3 course. Serum folic acid and fexofenadine concentrations were quantified with a chemiluminescence immunoassay and LC-MS/MS, respectively. Pharmacokinetics were compared between periods with standard bioequivalence approaches. RESULTS: While geometric mean folic acid AUC0-2h, which mainly reflects absorption, was 0.403 and 0.414 mg/L·h before and after the vitamin D3 course (geometric mean ratio (GMR), 1.027; 90 % confidence interval (90 % CI), 0.788-1.340), the geometric mean fexofenadine AUC0-2h was 1.932 and 2.761 mg/L·h, respectively (GMR, 1.429; 90 % CI, 0.890-2.294). PCFT- and OATP1A2-mRNA expressions in duodenal biopsies were essentially unchanged. CONCLUSIONS: No significant changes in folic acid and fexofenadine absorption were observed after a 10-day course of 1,25(OH)2D3 in humans in vivo. This study underlines the importance of confirming in vitro findings in vivo in humans.


Subject(s)
Folic Acid/pharmacokinetics , Intestinal Absorption/drug effects , Terfenadine/analogs & derivatives , Vitamin D/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Administration, Oral , Adult , Duodenum/drug effects , Duodenum/metabolism , Female , Folic Acid/blood , Healthy Volunteers , Humans , Male , Multidrug Resistance-Associated Proteins/genetics , Organic Anion Transporters/genetics , Proton-Coupled Folate Transporter/genetics , RNA, Messenger/metabolism , Terfenadine/blood , Terfenadine/pharmacokinetics , Vitamin D/pharmacology , Young Adult
19.
Biomed Chromatogr ; 30(7): 1059-1064, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26577375

ABSTRACT

A simple and sensitive method was developed for fexofenadine determination in human plasma by liquid chromatography with ultraviolet detection. Satisfactory separation was achieved on a Hypersil® BDS C18 column (250 × 4.6 mm, 5µm) using a mobile phase comprising 20 mm sodium dihydrogen phosphate-2 hydrate (pH adjusted to 3 with phosphoric acid)-acetonitrile at a ratio of 52:48, v/v. The elution was isocratic at ambient temperature with a flow rate of 1.0 mL/min. The UV detector was set at 215 nm for the drug and 330 nm for the internal standared (tinidazole). The total time for a chromatographic separation was ~6.5 min. Linearity was demonstrated over the concentration range 0.01-4 µg/mL. The observed within- and between-day assay precision ranged from 0.346 to 13.6%; accuracy varied between 100.4 and 111.2%. This method was successfully applied for therapeutic drug monitoring in patients treated with clinical doses of fexofenadine and for pharmacokinetic studies. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Chromatography, High Pressure Liquid/methods , Drug Monitoring/methods , Histamine H1 Antagonists, Non-Sedating/blood , Terfenadine/analogs & derivatives , Adult , Calibration , Histamine H1 Antagonists, Non-Sedating/pharmacokinetics , Humans , Quality Control , Terfenadine/blood , Terfenadine/pharmacokinetics , Young Adult
20.
Drug Dev Ind Pharm ; 42(6): 945-57, 2016.
Article in English | MEDLINE | ID: mdl-26467209

ABSTRACT

BACKGROUND: In the present age of polypharmacy, limited sampling strategy becomes important to verify if drug levels are within the prescribed threshold limits from efficacy and safety considerations. The need to establish reliable single time concentration dependent models to predict exposure becomes important from cost and time perspectives. METHODS: A simple unweighted linear regression model was developed to describe the relationship between Cmax versus AUC for fexofenadine, losartan, EXP3174, itraconazole and hydroxyitraconazole. The fold difference, defined as the quotient of the observed and predicted AUC values, were evaluated along with statistical comparison of the predicted versus observed values. RESULTS: The correlation between Cmax versus AUC was well established for all the five drugs with a correlation coefficient (r) ranging from 0.9130 to 0.9997. Majority of the predicted values for all the five drugs (77%) were contained within a narrow boundary of 0.75- to 1.5-fold difference. The r values for observed versus predicted AUC were 0.9653 (n = 145), 0.8342 (n = 76), 0.9524 (n = 88), 0.9339 (n = 89) and 0.9452 (n = 66) for fexofenadine, losartan, EXP3174, itraconazole and hydroxyitraconazole, respectively. CONCLUSIONS: Cmax versus AUC relationships were established for all drugs and were amenable for limited sampling strategy for AUC prediction. However, fexofenadine, EXP3174 and hydroxyitraconazole may be most relevant for AUC prediction by a single time concentration as judged by the various criteria applied in this study.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/pharmacokinetics , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP3A/metabolism , Itraconazole/pharmacokinetics , Losartan/pharmacokinetics , Terfenadine/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Area Under Curve , Cross-Over Studies , Drug Interactions , Humans , Itraconazole/analogs & derivatives , Itraconazole/metabolism , Losartan/metabolism , Polypharmacy , Terfenadine/metabolism , Terfenadine/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL