Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Clin Pharmacol ; 64(3): 371-377, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37751442

ABSTRACT

Hydrocephalus is a disorder caused by excess fluid accumulation in the brain and results in brain damage with consequent cognitive and physical problems. This condition has no cure; the only treatment is brain surgery. Experimental data indicate that P-glycoprotein (P-gp) plays a crucial role in the pathogenesis of hydrocephalus due to its function in clearing macromolecules from the brain. Numerous medications frequently used are classified as P-gp inducers or inhibitors, and comprehending their effects may aid in attaining improved patient outcomes. Therefore, in this single-center retrospective study, we examined the risk of the need for ventriculoperitoneal shunt placement over 10 years among 4588 adult patients with hydrocephalus not exposed to any P-gp inhibitors/inducers or exclusively exposed to either P-gp inhibitors or inducers. Our analysis shows that patients exposed to P-gp inhibitors had a 3.2 times higher risk of requiring ventriculoperitoneal shunt surgery (P < .0001). In contrast, the relative risk was not significantly affected (P = .07) among those exposed to P-gp inducers. Our findings indicate the need for caution when prescribing P-gp inhibitors to patients with hydrocephalus. Additional studies using larger cohorts are required to confirm whether P-gp inducers in patients with hydrocephalus can mitigate the risk of ventriculoperitoneal shunt.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1 , Hydrocephalus , Ventriculoperitoneal Shunt , Adult , Humans , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Hydrocephalus/etiology , Hydrocephalus/surgery , Retrospective Studies , Treatment Outcome
2.
CPT Pharmacometrics Syst Pharmacol ; 10(12): 1485-1496, 2021 12.
Article in English | MEDLINE | ID: mdl-34729944

ABSTRACT

Rifampicin induces both P-glycoprotein (P-gp) and cytochrome P450 3A4 (CYP3A4) through regulating common nuclear receptors (e.g., pregnane X receptor). The interplay of P-gp and CYP3A4 has emerged to be an important factor in clinical drug-drug interactions (DDIs) with P-gp-CYP3A4 dual substrates and requires qualitative and quantitative understanding. Although physiologically based pharmacokinetic (PBPK) modeling has become a widely accepted approach to assess DDIs and is able to reasonably predict DDIs caused by CYP3A4 induction and P-gp induction individually, the predictability of PBPK models for the effect of simultaneous P-gp and CYP3A4 induction on P-gp-CYP3A4 dual substrates remains to be systematically evaluated. In this study, we used a PBPK modeling approach for the assessment of DDIs between rifampicin and 12 drugs: three sensitive P-gp substrates, seven P-gp-CYP3A4 dual substrates, and two P-gp-CYP3A4 dual substrates and inhibitors. A 3.5-fold increase of intestinal P-gp abundance was incorporated in the PBPK models to account for rifampicin-mediated P-gp induction at steady state. The simulation results showed that accounting for P-gp induction in addition to CYP3A4 induction improved the prediction accuracy of the area under the concentration-time curve and maximum (peak) plasma drug concentration ratios compared with considering CYP3A4 induction alone. Furthermore, the interplay of relevant drug-specific parameters and its impact on the magnitude of DDIs were evaluated using sensitivity analysis. The PBPK approach described herein, in conjunction with robust in vitro and clinical data, can help in the prospective assessment of DDIs involving other P-gp and CYP3A4 dual substrates. The database reported in the present study provides a valuable aid in understanding the combined effect of P-gp and CYP3A4 induction during drug development.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , Cytochrome P-450 CYP3A Inducers/pharmacology , Models, Biological , Rifampin/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/pharmacokinetics , Computer Simulation , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP3A Inducers/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Interactions , Humans
3.
Daru ; 29(1): 13-22, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33405191

ABSTRACT

BACKGROUND: P-glycoprotein (P-gp) is an Adenosine triphosphate (ATP) dependent drug-efflux pump which is located abundantly in the stomach and protects the gut mucosa from xenobiotic. OBJECTIVE: The purpose of this study was to investigate the influence of P-gp modulation on the efficacy of treatment regimen. METHOD: P-gp modulation in rats was performed by using P-gp inducer (150 mg/kg rifampicin) and P-gp inhibitor (10 mg/kg cyclosporine A) for 14 days prior to be infected with Helicobacter pylori (H. pylori). The rats were further divided into groups, which were normal control, vehicle control, antibiotics and omeprazole, antibiotics only and omeprazole only for another 2 weeks of treatment. The ulcer formation and P-gp expression were determined by using macroscopic evaluation and western blot analysis, respectively. RESULTS: The highest P-gp expression was shown in the induced P-gp rats (2.00 ± 0.68) while the lowest P-gp expression was shown in the inhibited P-gp rats (0.45 ± 0.36) compared to the normal P-gp rats. In all groups, the rats which were infected with H. pylori, had a significant increase (p < 0.05) in P-gp expression level and a more severe ulcer formation compared to the healthy rats. The ulcer developed at different levels in the rats with inhibited, induced, or normal P-gp expression. After receiving the standard therapy for H. pylori, it was observed that the healing rate for ulcer was increased to 91% (rats with inhibited P-gp expression), 82% (rats with induced P-gp expression) and 75% in rats with normal P-gp. The use of rifampicin to induce P-gp level was also shown to be effective in eradicating the H. pylori infection. CONCLUSION: The synergism in the standard therapy by using two antibiotics (clarithromycin and amoxicillin) and proton pump inhibitor (omeprazole) have shown to effectively eradicate the H. pylori infection. Thus, P-gp expression influenced the effectiveness of the treatment.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Amoxicillin/therapeutic use , Anti-Bacterial Agents/therapeutic use , Anti-Ulcer Agents/therapeutic use , Clarithromycin/therapeutic use , Helicobacter Infections/drug therapy , Omeprazole/therapeutic use , Proton Pump Inhibitors/therapeutic use , Stomach Ulcer/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Cyclosporine/pharmacology , Drug Therapy, Combination , Helicobacter Infections/metabolism , Helicobacter pylori , Male , Rats, Sprague-Dawley , Rifampin/pharmacology , Stomach Ulcer/metabolism
4.
Food Chem Toxicol ; 146: 111785, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33011351

ABSTRACT

The drug transporter P-glycoprotein (P-gp) is often investigated in drug-interaction studies because the activity is modulated by a wide variety of xenobiotics including drugs, herbal products, and food components. In this study, we tested six common arylsulfonate food dyes-allura red, carmoisine, ponceau 4R, quinolone yellow, sunset yellow, and tartrazine-as activators and inhibitors of P-gp activity in vitro. The dyes were studied as P-gp activators by measuring ATPase activity in P-gp-expressing membranes. Compared to verapamil, a known activator of P-gp, the six food dyes showed no stimulatory activity. The potential for these six food dyes to act as P-gp inhibitors was tested in an intracellular efflux assay with P-gp-expressing cells. Compared to GF120918, a known P-gp inhibitor, there was no inhibitory activity for these six food dyes. The six food dyes tested do not interact with P-gp in vitro and, therefore, are unlikely cause clinical drug-food dye interactions. Further investigation is necessary to determine whether these food dyes could interact with other drug transporters.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Food Coloring Agents/pharmacology , Adenosine Triphosphatases/metabolism , Biological Transport , Drug Interactions , Food Coloring Agents/chemistry , Food-Drug Interactions , Humans , Verapamil/pharmacology
5.
J Clin Pharmacol ; 60(12): 1606-1616, 2020 12.
Article in English | MEDLINE | ID: mdl-33045114

ABSTRACT

Elagolix is an oral gonadotropin-releasing hormone receptor antagonist indicated for the management of endometriosis-associated pain and in combination with estradiol/norethindrone acetate indicated for the management of heavy menstrual bleeding associated with uterine leiomyomas (fibroids) in premenopausal women. Elagolix coadministered with estradiol/norethindrone acetate is in late-stage development for the management of heavy menstrual bleeding associated with uterine fibroids. Based on the in vitro profile of elagolix metabolism and disposition, 9 drug-drug interaction (DDI) studies evaluating the victim and perpetrator characteristics of elagolix were conducted in 144 healthy volunteers. As a victim of cytochrome P450 (CYPs) and transporter-mediated DDIs, elagolix area under the curve (AUC) increased by ∼2-fold following coadministration with ketoconazole and by ∼5- and ∼2-fold with single and multiple doses of rifampin, respectively. As a perpetrator, elagolix decreased midazolam AUC (90% confidence interval) by 54% (50%-59%) and increased digoxin AUC by 32% (23%-41%). Elagolix decreased rosuvastatin AUC by 40% (29%-50%). No clinically significant changes in exposure on coadministration with sertraline or fluconazole occurred. A elagolix 150-mg once-daily regimen should be limited to 6 months with strong CYP3A inhibitors and rifampin because of the potential increase in bone mineral density loss, as described in the drug label. A 200-mg twice-daily regimen is recommended for no more than 1 month with strong CYP3A inhibitors and not recommended with rifampin. Elagolix is contraindicated with strong organic anion transporter polypeptide B1 inhibitors (eg, cyclosporine and gemfibrozil). Consider increasing the doses of midazolam and rosuvastatin when coadministered with elagolix, and individualize therapy based on patient response. Clinical monitoring is recommended for P-glycoprotein substrates with a narrow therapeutic window (eg, digoxin). Dose adjustments are not required for sertraline, fluconazole, bupropion (or any CYP2B6 substrate), or elagolix when coadministered.


Subject(s)
Hydrocarbons, Fluorinated/administration & dosage , Hydrocarbons, Fluorinated/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Receptors, LHRH/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Adult , Cytochrome P-450 CYP2B6/metabolism , Cytochrome P-450 CYP2B6 Inducers/administration & dosage , Cytochrome P-450 CYP2B6 Inducers/pharmacokinetics , Cytochrome P-450 CYP2C9 Inhibitors/administration & dosage , Cytochrome P-450 CYP2C9 Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inducers/administration & dosage , Cytochrome P-450 CYP3A Inducers/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Drug Administration Schedule , Drug Interactions , Female , Healthy Volunteers , Humans , Hydrocarbons, Fluorinated/blood , Hydrocarbons, Fluorinated/pharmacology , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Liver-Specific Organic Anion Transporter 1/metabolism , Male , Middle Aged , Neoplasm Proteins/metabolism , Premenopause , Pyrimidines/blood , Pyrimidines/pharmacology , Solute Carrier Organic Anion Transporter Family Member 1B3/antagonists & inhibitors , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Young Adult
6.
Drugs ; 80(11): 1065-1083, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32504376

ABSTRACT

Edoxaban, a direct factor Xa inhibitor, is the latest of the non-vitamin K antagonist oral anticoagulants (NOACs). Despite being marketed later than other NOACs, its use is now spreading in current clinical practice, being indicated for both thromboprophylaxis in patients with non-valvular atrial fibrillation (NVAF) and for the treatment and prevention of venous thromboembolism (VTE). In patients with multiple conditions, the contemporary administration of several drugs can cause relevant drug-drug interactions (DDIs), which can affect drugs' pharmacokinetics and pharmacodynamics. Usually, all the NOACs are considered to have significantly fewer DDIs than vitamin K antagonists; notwithstanding, this is actually not true, all of them are affected by DDIs with drugs that can influence the activity (induction or inhibition) of P-glycoprotein (P-gp) and cytochrome P450 3A4, both responsible for the disposition and metabolism of NOACs to a different extent. In this review/expert opinion, we focused on an extensive report of edoxaban DDIs. All the relevant drugs categories have been examined to report on significant DDIs, discussing the impact on edoxaban pharmacokinetics and pharmacodynamics, and the evidence for dose adjustment. Our analysis found that, despite a restrained number of interactions, some strong inhibitors/inducers of P-gp and drug-metabolising enzymes can affect edoxaban concentration, just as it happens with other NOACs, implying the need for a dose adjustment. However, our analysis of edoxaban DDIs suggests that given the small propensity for interactions of this agent, its use represents an acceptable clinical decision. Still, DDIs can be significant in certain clinical situations and a careful evaluation is always needed when prescribing NOACs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1 , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Interactions , Pyridines/pharmacology , Thiazoles/pharmacology , Thromboembolism/prevention & control , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Factor Xa Inhibitors/pharmacology , Medication Therapy Management , Thromboembolism/etiology
8.
Curr Drug Metab ; 21(4): 281-290, 2020.
Article in English | MEDLINE | ID: mdl-32394829

ABSTRACT

BACKGROUND: Maytenus ilicifolia is a Brazilian popular medicine commonly used to treat ulcer and gastritis. Despite the absence of toxicity regarding its consumption, possible interactions when co-administrated with conventional drugs, are unknown. OBJECTIVE: This study aimed to evaluate the effects of M. ilicifolia extracts on Cytochrome P450 3A (CYP3A) and P-glycoprotein (P-gp) activities. METHODS: The extracts were obtained by infusion (MI) or turbo-extraction using hydro-acetonic solvent (MT70). The content of polyphenols in each extract was determined. To assess the modulation of M. ilicifolia on P-gp activity, the uptake of fexofenadine (FEX) by Caco-2 cells was investigated in the absence or presence of MI or MT70. The effect on CYP3A activity was evaluated by the co-administration of midazolam (MDZ) with each extract in male Wistar rats. The pharmacokinetic parameters of the drug were determined and compared with those from the control group. The content of total phenolic compounds, tannins, and flavonoids on MT70 extract was about double of that found in MI. RESULTS: In the presence of the extracts, the uptake of the P-gp marker (FEX) by Caco-2 cells increased from 1.7 ± 0.4 ng.mg-1 protein (control) to 3.5 ± 0.2 ng.mg-1 protein (MI) and 4.4 ± 0.5 ng.mg-1 protein (MT70), respectively. When orally co-administrated with MDZ (substrate of CYP3A), the extracts augmented the AUC(0-∞) (Control: 911.7 ± 215.7 ng.h.mL-1; MI: 1947 ± 554.3 ng.h.mL-1; MT70: 2219.0 ± 506.3 ng.h.mL-1) and the Cmax (Control: 407.7 ± 90.4 ng.mL-1; MI: 1770.5 ± 764.5 ng.mL-1; MT70: 1987.2 ± 544.9 ng.mL-1) of the drug in rats indicating a 50% reduction of the oral Cl. No effect was observed when midazolam was given intravenously. CONCLUSION: The results suggest that M. ilicifolia can inhibit the intestinal metabolism and transport of drugs mediated by CYP3A and P-gp, respectively, however, the involvement of other transporters and the clinical relevance of such interaction still need to be clarified.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cytochrome P-450 CYP3A/metabolism , Maytenus/chemistry , Plant Extracts/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , Animals , Caco-2 Cells , Cell Line , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Interactions , Humans , Ketoconazole/pharmacology , Male , Midazolam/pharmacokinetics , Quinolines/pharmacology , Rats , Rats, Wistar , Terfenadine/analogs & derivatives
9.
Eur J Pharm Sci ; 134: 194-204, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-31047967

ABSTRACT

Several reports demonstrated that rifampicin affected pharmacokinetics of victim drugs following oral more than intravenous administration. We aimed to establish a semi-physiologically based pharmacokinetic (semi-PBPK) model involving both enzyme and transporter turnover to simultaneously predict pharmacokinetic interaction of rifampicin with oral versus intravenous substrates of cytochrome P450 (CYP) 3A4/P­glycoprotein (P-GP) in human. Rifampicin was chosen as the CYP3A /P-GP inducer. Thirteen victim drugs including P-GP substrates (digoxin and talinolol), CYP3A substrates (alfentanil, midazolam, nifedipine, ondansetron and oxycodone), dual substrates of CYP3A/P-GP (quinidine, cyclosporine A, tacrolimus and verapamil) and complex substrates (S-ketamine and tramadol) were chosen to investigate drug-drug interactions (DDIs) with rifampicin. Corresponding parameters were cited from literatures. Before and after multi-dose of oral rifampicin, the pharmacokinetic profiles of victim drugs for oral or intravenous administration to human were predicted using the semi-PBPK model and compared with the observed values. Contribution of both CYP3A and P-GP induction in intestine and liver by rifampicin to pharmacokinetic profiles of victim drugs was investigated. The predicted pharmacokinetic profiles of drugs before and after rifampicin administration accorded with the observations. The predicted pharmacokinetic parameters and DDIs were successful, whose fold-errors were within 2. It was consistent with observations that the DDIs of rifampicin with oral victim drugs were larger than those with intravenous victim drugs. DDIs of rifampicin with CYP3A or P-GP substrates following oral versus intravenous administration to human were successfully predicted using the developed semi-PBPK model.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , Cytochrome P-450 CYP3A Inducers/pharmacokinetics , Pharmaceutical Preparations/metabolism , Rifampin/pharmacokinetics , Administration, Intravenous , Administration, Oral , Computer Simulation , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP3A Inducers/administration & dosage , Drug Interactions , Humans , Models, Biological , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Pharmacokinetics , Rifampin/administration & dosage
10.
Bioorg Chem ; 83: 242-249, 2019 03.
Article in English | MEDLINE | ID: mdl-30390553

ABSTRACT

Poly(ADP-ribose)polymerase-1 inhibitor (PARPi) AZD2461 was designed to be a weak P-glycoprotein (P-gp) analogue of FDA approved olaparib. With this chemical property in mind, we utilized the AZD2461 ligand architecture to develop a CNS penetrant and PARP-1 selective imaging probe, in order to investigate PARP-1 mediated neuroinflammation and neurodegenerative diseases, such as Alzheimer's and Parkinson's. Our work led to the identification of several high-affinity PARPi, including AZD2461 congener 9e (PARP-1 IC50 = 3.9 ±â€¯1.2 nM), which was further evaluated as a potential 18F-PET brain imaging probe. However, despite the similar molecular scaffolds of 9e and AZD2461, our studies revealed non-appreciable brain-uptake of [18F]9e in non-human primates, suggesting AZD2461 to be non-CNS penetrant.


Subject(s)
Blood-Brain Barrier/drug effects , Phthalazines/pharmacology , Piperidines/pharmacology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Cell Line, Tumor , Fluorine Radioisotopes/chemistry , Humans , Macaca mulatta , Male , Mice, Inbred BALB C , Phthalazines/chemical synthesis , Piperidines/chemical synthesis
11.
Biochem Pharmacol ; 154: 118-126, 2018 08.
Article in English | MEDLINE | ID: mdl-29684377

ABSTRACT

ABC transporters are key players in drug excretion with alterations in their expression and activity by therapeutic agents potentially leading to drug-drug interactions. The interaction potential of nomegestrol acetate (NMGA), a synthetic progestogen increasingly used as oral contraceptive, had never been explored. In this work we evaluated (1) the effect of NMGA on ABC transporters in the human hepatic cell line HepG2 and (2) the underlying molecular mechanism. NMGA (5, 50 and 500 nM) increased P-glycoprotein (P-gp) expression at both protein and mRNA levels and reduced intracellular calcein accumulation, indicating an increase also in transporter activity. This up-regulation of P-gp was corroborated in Huh7 cells and was independent of the classical progesterone receptor. Instead, using a siRNA-mediated silencing approach, we demonstrated the involvement of membrane progesterone receptor α. Moreover, we found that the activation of this receptor by NMGA led to a falling-rising profile in intracellular cAMP levels and protein kinase A activity over time, ultimately leading to transcriptional P-gp up-regulation. Finally, we identified inhibitory G protein and phosphodiesterases as mediators of this novel biphasic modulation. These results demonstrate the ability of NMGA to selectively up-regulate hepatic P-gp expression and activity and constitute the first report of ABC transporter modulation by membrane progesterone receptor α. If a similar regulation took place in vivo, decreased bioavailability and therapeutic efficacy of NMGA-coadministered P-gp substrates could be expected. This holds special importance considering long-term administration of NMGA and broad substrate specificity of P-gp.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Contraceptive Agents/pharmacology , Cyclic AMP/metabolism , Hepatocytes/metabolism , Megestrol/pharmacology , Norpregnadienes/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , Cyclic AMP/antagonists & inhibitors , Dose-Response Relationship, Drug , Gene Expression , Hep G2 Cells , Hepatocytes/drug effects , Humans
12.
Clin Pharmacol Ther ; 104(6): 1182-1190, 2018 12.
Article in English | MEDLINE | ID: mdl-29569723

ABSTRACT

Drug transporter and cytochrome P450 expression is regulated by shared nuclear receptors and, hence, an inducer should induce both, although the magnitude may differ. The objective of this study was to establish relative induction relationships between CYP3A and drug transporters (P-glycoprotein (P-gp), organic anion transporting polypeptide (OATP), and breast cancer resistance protein (BCRP)) or other P450s (CYP2C9 and CYP1A2) using ascending doses of the prototypical pregnane xenobiotic receptor (PXR) agonist, rifampin, to elicit weak, moderate, and strong PXR agonism. Healthy subjects received dabigatran etexilate, pravastatin, rosuvastatin, and a midazolam/tolbutamide/caffeine cocktail before and after rifampin 2, 10, 75, or 600 mg q.d. Unlike CYP3A, only moderate induction of P-gp, OATP, and CYP2C9 was observed and dose-dependent induction of P-gp, OATP, and CYP2C9 was always one drug-drug interaction category lower than observed for CYP3A, even when correcting for probe drug sensitivity. Data from this study establish proof-of-concept that P450 induction data can be leveraged to inform on the effect on transporters.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , Cytochrome P-450 CYP3A/biosynthesis , Cytochrome P-450 Enzyme Inducers/administration & dosage , Membrane Transport Modulators/administration & dosage , Pregnane X Receptor/agonists , Rifampin/administration & dosage , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adolescent , Adult , Biotransformation , Computer Simulation , Cytochrome P-450 Enzyme Inducers/adverse effects , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Induction , Female , Healthy Volunteers , Humans , Male , Membrane Transport Modulators/adverse effects , Middle Aged , Models, Biological , Organic Anion Transporters/agonists , Organic Anion Transporters/metabolism , Pharmacokinetics , Pregnane X Receptor/metabolism , Rifampin/adverse effects , Risk Assessment , Substrate Specificity , Young Adult
13.
Clin Pharmacol Ther ; 104(6): 1191-1198, 2018 12.
Article in English | MEDLINE | ID: mdl-29569712

ABSTRACT

Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. The objective of this study was to determine if these relationships could be utilized to predict transporter induction by other CYP3A inducers (rifabutin and carbamazepine) and of another P-gp substrate, sofosbuvir. Healthy subjects received sofosbuvir and a six-probe drug cassette before and after 300 mg q.d. rifabutin or 300 mg b.i.d. carbamazepine. Induction of P-gp, CYP2C9, and decreased sofosbuvir exposure were successfully predicted by observed CYP3A induction. Carbamazepine induction of OATP was underpredicted, likely due to reported additional non-PXR agonism. The results demonstrate that the effect of a PXR agonist on CYP3A can be leveraged to inform on induction liability for other primarily PXR-regulated P450s/transporters, allowing for prioritization of targeted DDI assessments during new drug development.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , Carbamazepine/administration & dosage , Cytochrome P-450 CYP3A/biosynthesis , Cytochrome P-450 Enzyme Inducers/administration & dosage , Membrane Transport Modulators/administration & dosage , Pregnane X Receptor/agonists , Rifabutin/administration & dosage , Rifampin/administration & dosage , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adolescent , Adult , Biotransformation , Carbamazepine/adverse effects , Computer Simulation , Cytochrome P-450 CYP2C9/biosynthesis , Cytochrome P-450 Enzyme Inducers/adverse effects , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Induction , Female , Healthy Volunteers , Humans , Male , Membrane Transport Modulators/adverse effects , Middle Aged , Models, Biological , Organic Anion Transporters/agonists , Organic Anion Transporters/metabolism , Pregnane X Receptor/metabolism , Rifabutin/adverse effects , Rifampin/adverse effects , Risk Assessment , Sofosbuvir/metabolism , Substrate Specificity , Young Adult
14.
Eur J Drug Metab Pharmacokinet ; 43(5): 533-541, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29500603

ABSTRACT

BACKGROUND: Nintedanib is a substrate for p-glycoprotein which can impact bioavailability. We investigated the effects of ketoconazole, a p-glycoprotein inhibitor, and rifampicin, a p-glycoprotein inducer, on the pharmacokinetics of nintedanib. METHODS: In the ketoconazole study, 34 healthy subjects received nintedanib 50 mg orally alone and 1 h after the last dose of ketoconazole given orally at a dose of 400 mg once daily for 3 days in 1 of 2 randomized sequences. In the rifampicin study, 26 subjects received nintedanib 150 mg orally alone and the morning after the last dose of rifampicin given orally at a dose of 600 mg once daily for 7 days. The primary objective was to determine the relative bioavailability of nintedanib administered following multiple doses of ketoconazole or rifampicin versus alone, based on AUC from time 0 extrapolated to infinity (AUC0-∞) and maximum concentration (Cmax) calculated using an analysis of variance. Geometric mean ratios and 2-sided 90% CIs were calculated. RESULTS: Exposure to nintedanib increased when it was administered following ketoconazole versus alone (AUC0-∞: geometric mean ratio, 160.5% [90% CI, 148.2-173.7]; Cmax: geometric mean ratio, 179.6% [90% CI, 157.6-204.8]) and decreased when it was administered following rifampicin versus alone (AUC0-∞: geometric mean ratio, 50.1% [90% CI, 47.2-53.3]; Cmax: geometric mean ratio, 59.8% [90% CI, 53.8-66.4]). The time to reach Cmax (tmax) and half-life (t½) of nintedanib were unaffected by co-administration of ketoconazole or rifampicin. CONCLUSIONS: Exposure to nintedanib is increased by co-administration of ketoconazole and decreased by co-administration of rifampicin, likely due to effects on bioavailability of the absorbed fraction. ClinicalTrials.govidentifiers:NCT01679613, NCT01770392.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Indoles/administration & dosage , Indoles/pharmacokinetics , Ketoconazole/administration & dosage , Rifampin/administration & dosage , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Administration, Oral , Adolescent , Adult , Biological Availability , Cross-Over Studies , Drug Administration Schedule , Drug Interactions , Germany , Healthy Volunteers , Humans , Indoles/adverse effects , Intestinal Absorption , Ketoconazole/adverse effects , Male , Middle Aged , Models, Biological , Rifampin/adverse effects , Risk Assessment , Young Adult
15.
Clin Pharmacokinet ; 56(3): 235-250, 2017 03.
Article in English | MEDLINE | ID: mdl-27470518

ABSTRACT

Afatinib is an oral, irreversible ErbB family blocker that covalently binds to the kinase domains of epidermal growth factor receptor (EGFR), human EGFRs (HER) 2, and HER4, resulting in irreversible inhibition of tyrosine kinase autophosphorylation. Studies in healthy volunteers and patients with advanced solid tumours have shown that once-daily afatinib has time-independent pharmacokinetic characteristics. Maximum plasma concentrations of afatinib are reached approximately 2-5 h after oral administration and thereafter decline, at least bi-exponentially. Food reduces total exposure to afatinib. Over the clinical dose range of 20-50 mg, afatinib exposure increases slightly more than dose proportional. Afatinib metabolism is minimal, with unchanged drug predominantly excreted in the faeces and approximately 5 % in urine. Apart from the parent drug afatinib, the major circulation species in human plasma are the covalently bound adducts to plasma protein. The effective elimination half-life is approximately 37 h, consistent with an accumulation of drug exposure by 2.5- to 3.4-fold based on area under the plasma concentration-time curve (AUC) after multiple dosing. The pharmacokinetic profile of afatinib is consistent across a range of patient populations. Age, ethnicity, smoking status and hepatic function had no influence on afatinib pharmacokinetics, while females and patients with low body weight had increased exposure to afatinib. Renal function is correlated with afatinib exposure, but, as for sex and body weight, the effect size for patients with severe renal impairment (approximately 50 % increase in AUC) is only mildly relative to the extent of unexplained interpatient variability in afatinib exposure. Afatinib has a low potential as a victim or perpetrator of drug-drug interactions, especially with cytochrome P450-modulating agents. However, concomitant treatment with potent inhibitors or inducers of the P-glycoprotein transporter can affect the pharmacokinetics of afatinib. At a dose of 50 mg, afatinib does not have proarrhythmic potential.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Neoplasms/metabolism , Quinazolines/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Afatinib , Animals , Drug Interactions/physiology , Humans , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Neoplasms/drug therapy , Protein Binding/physiology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Radiation-Sensitizing Agents/pharmacokinetics , Radiation-Sensitizing Agents/therapeutic use
16.
Adv Clin Exp Med ; 25(2): 237-42, 2016.
Article in English | MEDLINE | ID: mdl-27627555

ABSTRACT

BACKGROUND: Permeability glycoprotein (P-glycoprotein or P-gp) plays an important role in the intestinal absorption of the immunosuppressive agents: cyclosporine and tacrolimus. OBJECTIVES: The aim of this study was to determine how the intestinal absorption of cyclosporine and tacrolimus is affected when they are used with P-gp activating or inhibiting agents. MATERIAL AND METHODS: In in vitro experiments, everted parts of rat small intestines were used to evaluate the effects of verapamil (a P-gp inhibitor) and rifampicin (a P-gp inducer) on the intestinal absorption of cyclosporine and tacrolimus. In in vivo experiments, the effects of verapamil and rifampicin on the plasma concentrations of cyclosporine and tacrolimus were evaluated. RESULTS: In in vitro experiments, the absorption of cyclosporine and tacrolimus from the small intestine increased in a time-dependent manner when the drugs were administered with or without verapamil or rifampicin. There was no difference in the absorption of cyclosporine ± verapamil/rifampicin between the jejunum and ileum; however, ileal absorption of tacrolimus + rifampicin was significantly higher than jejunal absorption (p < 0.05). Plasma concentrations of cyclosporine and tacrolimus were significantly increased when they were co-administered with verapamil (p < 0.001) and significantly decreased when co-administered with rifampicin (p < 0.05). CONCLUSIONS: P-gp may play an important role in the absorption of immunosupressive drugs, and it may contribute to drug-drug interactions thay may lead to inadequate drug response or toxicity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Cyclosporine/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Intestinal Absorption/drug effects , Intestine, Small/drug effects , Rifampin/pharmacology , Tacrolimus/pharmacokinetics , Verapamil/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Administration, Oral , Animals , Cyclosporine/administration & dosage , Cyclosporine/blood , Drug Interactions , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/blood , In Vitro Techniques , Intestine, Small/metabolism , Male , Rats, Sprague-Dawley , Rifampin/administration & dosage , Tacrolimus/administration & dosage , Tacrolimus/blood , Verapamil/administration & dosage
17.
Biochem Pharmacol ; 96(3): 190-201, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26070251

ABSTRACT

A number of statins, the cholesterol-lowering drugs, inhibit the in vitro replication of hepatitis C virus (HCV). In HCV-infected patients, addition of statins to the earlier standard of care therapy (pegIFN-α and ribavirin) resulted in increased sustained virological response rates. The mechanism by which statins inhibit HCV replication has not yet been elucidated. In an attempt to gain insight in the underlying mechanism, hepatoma cells carrying an HCV replicon were passaged in the presence of increasing concentrations of fluvastatin. Fluvastatin-resistant replicon containing cells could be generated and proved ∼8-fold less susceptible to fluvastatin than wild-type cultures. The growth efficiency of the resistant replicon containing cells was comparable to that of wild-type replicon cells. The fluvastatin-resistant phenotype was not conferred by mutations in the viral genome but is caused by cellular changes. The resistant cell line had a markedly increased HMG-CoA reductase expression upon statin treatment. Furthermore, the expression of the efflux transporter P-gp was increased in fluvastatin-resistant replicon cells (determined by qRT-PCR and flow cytometry). This increased expression resulted also in an increased functional transport activity as measured by the P-gp mediated efflux of calcein AM. In conclusion, we demonstrate that statin resistance in HCV replicon containing hepatoma cells is conferred by changes in the cellular environment.


Subject(s)
Anticholesteremic Agents/pharmacology , Antiviral Agents/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Hepacivirus/drug effects , Host-Pathogen Interactions , Indoles/pharmacology , Virus Replication/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Carbamates , Cell Line, Tumor , Drug Resistance, Viral , Enzyme Activation , Fluvastatin , Gene Expression Regulation , Hepacivirus/genetics , Hepacivirus/growth & development , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/virology , Humans , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/genetics , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Imidazoles/pharmacology , Oligopeptides/pharmacology , Pyrrolidines , Replicon , Signal Transduction , Valine/analogs & derivatives , Viral Proteins/antagonists & inhibitors , Viral Proteins/genetics , Viral Proteins/metabolism
18.
Tsitologiia ; 56(11): 841-9, 2014.
Article in Russian | MEDLINE | ID: mdl-25707211

ABSTRACT

In this article we show that long-term cultivation of Chinese hamster fibroblasts of line V-79 RJK at elevated temperature resulted in the selection of variants with genetic changes at the level of karyotype. From the first steps of resistance selection to elevated temperature we identified population of cells with changes in karyotype (polyploidy cells, deletions, inversions, translocations of chromosomes, and some cells with DM-chromosomes). Further cultivation was accompanied with selection of cells with paracentrical chromosome breakages and HSR's on chromosomes. Nonspecific destabilization of the karyotype (on first steps of selection) was associated with increased expression of hsc70 and pgp. After long-term incubation at an elevated temperature, the cells with karyotypic changes had the basal level of hsc70 and pgp expression.


Subject(s)
Chromosome Aberrations , Fibroblasts/metabolism , Genomic Instability , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Cell Line , Cricetulus , Fibroblasts/pathology , Gene Expression , HSP70 Heat-Shock Proteins/agonists , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/agonists , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Hot Temperature , Karyotype
19.
Antimicrob Agents Chemother ; 57(9): 4481-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23836171

ABSTRACT

The membrane-associated drug transporter P-glycoprotein (P-gp) plays an essential role in drug efflux from the brain. Induction of this protein at the blood-brain barrier (BBB) could further affect the ability of a drug to enter the brain. At present, P-gp induction mediated by antiretroviral drugs at the BBB has not been fully investigated. Since P-gp expression is regulated by ligand-activated nuclear receptors, i.e., human pregnane X receptor (hPXR) and human constitutive androstane receptor (hCAR), these receptors could represent potential pathways involved in P-gp induction by antiretroviral drugs. The aims of this study were (i) to determine whether antiretroviral drugs currently used in HIV pharmacotherapy are ligands for hPXR or hCAR and (ii) to examine P-gp function and expression in human brain microvessel endothelial cells treated with antiretroviral drugs identified as ligands of hPXR and/or hCAR. Luciferase reporter gene assays were performed to examine the activation of hPXR and hCAR by antiretroviral drugs. The hCMEC/D3 cell line, which is known to display several morphological and biochemical properties of the BBB in humans, was used to examine P-gp induction following 72 h of exposure to these agents. Amprenavir, atazanavir, darunavir, efavirenz, ritonavir, and lopinavir were found to activate hPXR, whereas abacavir, efavirenz, and nevirapine were found to activate hCAR. P-gp expression and function were significantly induced in hCMEC/D3 cells treated with these drugs at clinical concentrations in plasma. Together, our data suggest that P-gp induction could occur at the BBB during chronic treatment with antiretroviral drugs identified as ligands of hPXR and/or hCAR.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , Antiviral Agents/pharmacology , Endothelial Cells/drug effects , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Steroid/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain/blood supply , Brain/cytology , Brain/drug effects , Brain/metabolism , Cell Line , Constitutive Androstane Receptor , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Microvessels/cytology , Microvessels/drug effects , Microvessels/metabolism , Pregnane X Receptor , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Signal Transduction
20.
Cancer Chemother Pharmacol ; 72(1): 189-99, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23673445

ABSTRACT

PURPOSE: Specific tyrosine kinase inhibitors were recently reported to modulate the activity of ABC transporters, leading to an increase in the intracellular concentration of their substrate drugs. In this study, we determine whether PD173074, a specific fibroblast growth factor receptor (FGFR) inhibitor, could reverse ABC transporter-mediated multidrug resistance. METHODS: 3-(4,5-Dimethylthiazol-yl)-2,5-diphenyllapatinibrazolium bromide assay was used to determine the effect of PD173074 on reversal of ABC transporter-mediated multidrug resistance (MDR). In addition, [³H]-paclitaxel accumulation/efflux assay, western blotting analysis, ATPase, and photoaffinity labeling assays were done to study the interaction of PD173074 on ABC transporters. RESULTS: PD173074 significantly sensitized both ABCB1-transfected and drug-selected cell lines overexpressing this transporter to substrate anticancer drugs colchicine, paclitaxel, and vincristine. This effect of PD173074 is specific to ABCB1, as no significant interaction was detected with other ABC transporters such as ABCC1 and ABCG2. The observed reversal effect seems to be primarily due to the decreased active efflux of [³H]-paclitaxel in ABCB1 overexpressing cells observed in efflux assay. In addition, no significant change in the ABCB1 expression was observed when ABCB1 overexpressing cells were exposed to 5 µM PD173074 for up to 3 days, thereby further suggesting its role in modulating the function of the transporter. In addition, PD173074 stimulated the ATPase activity of ABCB1 in a concentration-dependent manner, indicating a direct interaction with the transporter. Interestingly, PD173074 did not inhibit photolabeling of ABCB1 with [¹²5I]-iodoarylazidoprazosin (IAAP), showing that it binds at a site different from that of IAAP in the drug-binding pocket. CONCLUSIONS: Here, we report for the first time, PD173074, an inhibitor of the FGFR, to selectively reverse ABCB1 transporter-mediated MDR by directly blocking the efflux function of the transporter.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antineoplastic Agents/agonists , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenosine Triphosphate/metabolism , Affinity Labels/pharmacology , Allosteric Regulation , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Biological Transport/drug effects , Cell Line, Tumor , Colchicine/agonists , Colchicine/pharmacology , HEK293 Cells , Humans , Hydrolysis/drug effects , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Neoplasms/metabolism , Paclitaxel/agonists , Paclitaxel/metabolism , Paclitaxel/pharmacology , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Tubulin Modulators/agonists , Tubulin Modulators/metabolism , Tubulin Modulators/pharmacology , Vincristine/agonists , Vincristine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...