Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
2.
Br J Cancer ; 112(12): 1874-81, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-25989270

ABSTRACT

BACKGROUND: The mitogen-activated protein kinase (MAPK) pathway has been implicated in the molecular pathogenesis of human cancers, including metastatic colorectal cancer (mCRC). This provides a rationale for the development of MAPK-targeted agents such as pimasertib. METHODS: Patients with KRAS mutant mCRC were treated in the second-line setting with FOLFIRI (5-fluorouracil/folinic acid/irinotecan) plus pimasertib. The primary objective of the safety run-in phase was to determine the maximum-tolerated dose (MTD) and the recommended phase II dose of pimasertib combined with FOLFIRI. RESULTS: Sixteen patients were enrolled in the trial. Ten and six patients were treated daily with 45 and 60 mg of pimasertib plus FOLFIRI, respectively. The MTD was considered to be 45 mg per day. The most common treatment-emergent adverse events were diarrhoea, nausea, vomiting, asthenia and skin/rash event. Of the 15 patients in the efficacy analysis group, two patients had partial response, nine patients had stable disease, three patients had progressive disease as their best overall response and one patient could not be evaluated. CONCLUSIONS: Dose escalation of pimasertib in combination with FOLFIRI was limited by toxicity. At the MTD of 45 mg per day, pimasertib was adequately tolerated in patients with mCRC and no unexpected or new safety signals or concerns were identified.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Genes, ras , Humans , Leucovorin/administration & dosage , Leucovorin/pharmacokinetics , Male , Middle Aged , Mutation , Neoplasm Metastasis , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Niacinamide/pharmacokinetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Treatment Outcome , ras Proteins/genetics
3.
Xenobiotica ; 41(8): 712-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21657832

ABSTRACT

Transporter mediated drug-drug interactions (tDDI) mediated by ABCB1 have been shown to be clinically relevant. Hence, the assessment of the ABCB1 tDDI potential early in the drug development process has gained interest. We have evaluated the Calcein assay as a means of assessing the ABCB1 tDDI that is amenable to high throughout and compared it with the monolayer efflux assay. We found the Calcein assay, when performed in K562MDR cells using the protocol originally published more sensitive than digoxin transport inhibition in MDCKII-MDR1 cells. Application of the Calcein assay to cell lines containing different amounts of ABCB1, yielded IC(50) values that varied 10-100-fold. The differences observed for IC(50) values for the same compounds were in the following rank order: IC(50, MDCKII-MDR1) >IC(50, K562MDR)>IC(50, hCMEC/D3). Higher IC(50) values were obtained in cells with higher ABCB1 expression. The Calcein assay is a high-throughput alternative to digoxin transport inhibition as it appears to have a comparable selectivity but higher sensitivity than previously published digoxin transport inhibition in MDCKII-MDR1 cells. In addition, it can be performed in a barrier-specific manner highlighting the dependence of ABCB1 IC(50) values on different ABCB1 expression levels.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Fluoresceins/chemistry , Fluorescent Dyes/chemistry , High-Throughput Screening Assays , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Line , Drug Interactions , Humans , Sensitivity and Specificity
4.
Drug Metab Dispos ; 37(4): 794-801, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19118132

ABSTRACT

Multidrug resistance protein 2 (MRP2) is a multispecific organic anion transporter expressed at important pharmacological barriers, including the canalicular membrane of hepatocytes. At this location it is involved in the elimination of both endogenous and exogenous waste products, mostly as conjugates, to the bile. Estradiol-17beta-d-glucuronide (E(2)17betaG), a widely studied endogenous substrate of MRP2, was shown earlier to recognize two binding sites of the transporter in vesicular transport assays. MRP2 modulators (substrates and nonsubstrates) potentiate the transport of E(2)17betaG by MRP2. We correlated data obtained from studies of different complexities and investigated the species-specific differences between rat and human MRP2-mediated transport. We used vesicular transport assays, sandwich-cultured primary hepatocytes, and in vivo biliary efflux in rats. Our results demonstrate that the rat Mrp2 transporter, unlike the human MRP2, transports E(2)17betaG according to Michaelis-Menten type kinetics. Nevertheless, in the presence of modulator drugs E(2)17betaG transport mediated by the rat transporter also shows cooperative kinetics as potentiation of E(2)17betaG transport was observed in the vesicular transport assay. We also demonstrated that the potentiation exists both in rat and in human hepatocytes and in vivo in rats.


Subject(s)
Estradiol/analogs & derivatives , Multidrug Resistance-Associated Proteins/physiology , Animals , Biological Transport , Cells, Cultured , Estradiol/metabolism , Estradiol/pharmacokinetics , Hepatocytes/metabolism , Humans , Male , Multidrug Resistance-Associated Protein 2 , Rats , Rats, Wistar , Species Specificity
5.
J Pharmacol Exp Ther ; 321(3): 1085-94, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17347325

ABSTRACT

ABCG2, a transporter of the ATP-binding cassette family, is known to play a prominent role in the absorption, distribution, metabolism, and excretion of xenobiotics. Drug-transporter interactions are commonly screened by high-throughput systems using transfected insect and/or human cell lines. The determination of ABCG2-ATPase activity is one method to identify ABCG2 substrate and inhibitors. We demonstrate that the ATPase activities of the human ABCG2 transfected Sf9 cell membranes (MXR-Sf9) and ABCG2-overexpressing human cell membranes (MXR-M) differ. Variation due to disparity in the glycosylation level of the protein had no effect on the transporter. The influence of cholesterol on ABCG2-ATPase activity was investigated because the lipid compositions of insect and human cells are largely different from each other. Differences in cholesterol content, shown by cholesterol loading and depletion experiments, conferred the difference in stimulation of basal ABCG2-ATPase of the two cell membranes. Basal ABCG2-ATPase activity could be stimulated by sulfasalazine, prazosin, and topotecan, known substrates of ABCG2 in cholesterol-loaded MXR-Sf9 and MXR-M cell membranes. In contrast, ABCG2-ATPase could not be stimulated in MXR-Sf9 or in cholesterol-depleted MXR-M membranes. Moreover, cholesterol loading significantly improved the drug transport into inside-out membrane vesicles prepared from MXR-Sf9 cells. MXR-M and cholesterol-loaded MXR-Sf9 cell membranes displayed similar ABCG2-ATPase activity and vesicular transport. Our study indicates an essential role of membrane cholesterol for the function of ABCG2.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Cell Membrane/metabolism , Cholesterol/physiology , Neoplasm Proteins/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphatases/metabolism , Animals , Baculoviridae/genetics , Benzimidazoles/metabolism , Biological Transport, Active/drug effects , Cell Line , Cholesterol/pharmacology , Estrone/analogs & derivatives , Estrone/metabolism , Glycosylation , Humans , Kinetics , Methotrexate/metabolism , Neoplasm Proteins/genetics , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Pharmaceutical Preparations/metabolism , Prazosin/metabolism , Spodoptera , Sulfasalazine/metabolism , Topotecan/metabolism
6.
Clin Pharmacol Ther ; 70(3): 217-27, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11557909

ABSTRACT

BACKGROUND AND AIMS: The contribution of the gastrointestinal tract in comparison with the liver for the low and variable bioavailability of orally administered drugs is still poorly understood. Here we report on a new intestinal perfusion technique for the direct assessment of absorption, metabolism, and transport of drugs by the intestinal wall. METHODS: In 6 healthy volunteers a multilumen perfusion catheter was used to generate a 20-cm isolated jejunal segment that was perfused with 80 mg verapamil. Simultaneously, 5 mg [(2)H(7)]verapamil was given intravenously. Blood, perfusate, and bile samples were analyzed for parent verapamil and its major metabolites. RESULTS: The mean fraction of the verapamil dose absorbed from the 20-cm segment was 0.76 but substantial interindividual variability (0.51-0.96) was shown. Bioavailability was low (19.3%). The intestinal wall contributed to the same extent as the liver to extensive first-pass metabolism (mean extraction ratio, 0.49 versus 0.48). Substantial transport of verapamil metabolites from the systemic circulation via the enterocytes into the intestinal lumen was observed. Compared with biliary excretion, intestinal secretion into a 20-cm jejunal segment contributed to drug elimination to a similar extent. CONCLUSION: First-pass metabolism by the intestinal wall is extensive and contributes to the same extent as the liver to low bioavailability of some drugs such as verapamil. Moreover, intestinal secretion is as important as biliary excretion for the elimination of metabolites.


Subject(s)
Intestinal Absorption/physiology , Intestinal Mucosa/metabolism , Liver/metabolism , Pharmaceutical Preparations/metabolism , Adult , Algorithms , Area Under Curve , Bile/metabolism , Biological Availability , Biological Transport , Calcium Channel Blockers/pharmacokinetics , Humans , Male , Middle Aged , Perfusion , Verapamil/pharmacokinetics
7.
Biochem Biophys Res Commun ; 284(2): 455-60, 2001 Jun 08.
Article in English | MEDLINE | ID: mdl-11394901

ABSTRACT

Human cytochrome P450 2A6 (CYP2A6) constitutes the major nicotine oxidase, and large interindividual differences are seen in the levels of this enzyme, to a great extent caused by the distribution of several different polymorphic gene variants mainly located in the open reading frame (ORF). In the present study, we report a common polymorphism located in the 5' flanking region of CYP2A6 affecting its expression. DHPLC analysis and complete sequence of the open reading frame of the gene from a Turkish individual revealed a -48T > G substitution disrupting the TATA box. Using dynamic allele-specific hybridization (DASH), genotyping of this novel variant (named CYP2A6*9) was carried out in 116 Swedish, 132 Turkish, and 102 Chinese subjects, and the allele frequencies were found to be 5.2, 7.2, and 15.7%, respectively. The significance of the polymorphism was investigated by the construction of luciferase reporter plasmids containing 135 or 500 bp of the 5'-upstream region of the gene transfected into human hepatoma B16A2 cells. The constructs carrying the -48T > G mutation were only expressed at about 50% of the wild-type alleles. It is concluded that the CYP2A6*9 allele might be one of the most common CYP2A6 variants in Caucasians that alters the levels of enzyme expression.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/genetics , Mixed Function Oxygenases/genetics , Oxidoreductases/genetics , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/physiology , TATA Box/genetics , Alleles , Asian People/genetics , Cell Line , China/ethnology , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2A6 , DNA Mutational Analysis , Gene Expression/physiology , Genes, Reporter , Genetic Testing , Genetic Variation , Genotype , Humans , Nucleic Acid Denaturation , Nucleic Acid Heteroduplexes/analysis , Nucleic Acid Heteroduplexes/genetics , Nucleic Acid Hybridization/methods , Sweden/ethnology , Transfection , Turkey/ethnology , White People/genetics
8.
Clin Pharmacol Ther ; 68(4): 345-55, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11061574

ABSTRACT

BACKGROUND: P-Glycoprotein is an efflux pump in many epithelial cells with excretory function. It has been demonstrated that rifampin (INN, rifampicin) induces P-glycoprotein, particularly in the gut wall. We therefore hypothesized that rifampin affects pharmacokinetics of the P-glycoprotein substrate talinolol, a beta1-blocker without appreciable metabolic disposition but intense intestinal secretion in human beings. METHODS: Pharmacokinetics of talinolol (a single dose of 30 mg administered intravenously or 100 mg administered orally for 7 days) and duodenal expression of the MDR1 gene product P-glycoprotein as assessed by reverse transcriptase-polymerase chain reaction of the MDR1-messenger ribonucleic acid, by immunohistochemistry and Western blot analysis were analyzed before and after coadministration of rifampin (600 mg per day for 9 days) in 8 male healthy volunteers (age 22 to 26 years). RESULTS: During rifampin treatment, the areas under the curve of intravenous and oral talinolol were significantly lower (21% and 35%; P < .05). Treatment with rifampin resulted in a significantly increased expression of duodenal P-glycoprotein content 4.2-fold (2.9, 6.51) (Western blot) and messenger RNA was increased in six of the eight volunteers. P-Glycoprotein expression in biopsy specimens of gut mucosa correlated significantly with the systemic clearance of intravenous talinolol (rs = 0.74; P < .001). CONCLUSIONS: Rifampin induces P-glycoprotein-mediated excretion of talinolol predominantly in the gut wall. Moreover, clearance of talinolol from the blood into the lumen of the gastrointestinal tract may be predicted by the individual intestinal P-glycoprotein expression. Thus we describe a new type of steady-state drug interaction affecting compounds that are subject to transport rather than metabolism.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adrenergic beta-Antagonists/pharmacokinetics , Antitubercular Agents/pharmacology , Duodenum/metabolism , Propanolamines/pharmacokinetics , Rifampin/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Administration, Oral , Adrenergic beta-Antagonists/administration & dosage , Adrenergic beta-Antagonists/blood , Adult , Anti-Arrhythmia Agents/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Area Under Curve , Blotting, Western , Endoscopy, Digestive System , Enzyme Induction/drug effects , Humans , Immunohistochemistry , Infusions, Intravenous , Male , Propanolamines/administration & dosage , Propanolamines/blood , RNA, Messenger/analysis , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
9.
Drug Metab Dispos ; 28(10): 1149-52, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10997932

ABSTRACT

There is very limited knowledge about possible pharmacokinetic interactions between opioid analgesics and nonsteroidal antiinflammatory drugs (NSAIDs), which are commonly used in combination for the treatment of chronic pain. The major metabolic pathway of the weak opioid codeine is glucuronidation to codeine-6-glucuronide. Therefore we investigated the influence of the NSAID diclofenac on the formation of codeine-6-glucuronide in vitro, using human liver tissue homogenate. The formation of codeine-6-glucuronide exhibited single enzyme Michaelis-Menten kinetics with an average V(max) of 93.6 +/- 35.3 pmol/mg/min. A noncompetitive inhibition of codeine-6-glucuronidation by diclofenac was observed with an average K(i) of 7.9 microM. These in vitro findings suggest that a pharmacokinetic interaction occurs in vivo, which has to be confirmed by an interaction study in human subjects. It can be speculated that in case of inhibition of glucuronidation, the amount of codeine available for other pathways especially O-demethylation to morphine is increased, resulting in higher morphine serum levels and therefore higher analgesic efficacy.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Codeine/metabolism , Diclofenac/pharmacology , Narcotics/metabolism , Aged , Codeine/analogs & derivatives , Codeine/pharmacokinetics , Dose-Response Relationship, Drug , Drug Interactions , Humans , In Vitro Techniques , Intestine, Small/drug effects , Intestine, Small/metabolism , Kinetics , Liver/drug effects , Liver/metabolism , Male , Narcotics/pharmacokinetics
10.
J Chromatogr B Biomed Sci Appl ; 738(1): 137-47, 2000 Jan 28.
Article in English | MEDLINE | ID: mdl-10778935

ABSTRACT

A rapid and highly sensitive method for the determination of verapamil [2,8-bis-(3,4-dimethoxyphenyl)-6-methyl-2-isopropyl-6-azaoctanitrile+ ++] and [2H7]verapamil and their primary metabolites D-617 [2-(3,4-dimethoxyphenyl)-5-methylamino-2-isopropylvaleronitrile], D-703 [2-(4-hydroxy-3-methoxyphenyl)-8-(3,4-dimethoxy-phenyl)-6-methyl-2-iso-p ropyl-6-azaoctanitrile], D-702 [2-(3,4-dimethoxy-phenyl)-8-(4-hydroxy-3-methoxyphenyl)-6-methyl-2-isopr opyl-6-azaoctanitrile], norverapamil [2,8-bis-(3,4-dimethoxyphenyl)-2-isopropyl-6-azaoctanitrile] and secondary metabolites D-620 [2-(3,4-dimethoxyphenyl)-5-amino-2-isopropylvaleronitrile], D-717 [2-(4-hydroxy-3-methoxyphenyl)-5-amino-2-isopropylvaleronitrile], and D-715 [2-(4-hydroxy-3-methoxyphenyl)-8-(3,4-dimethoxy-phenyl)-2-isopropyl-6-++ +azaoctanitrile] has been developed using high-performance liquid chromatography-electrospray mass spectrometry. D-832, the gallopamil analogue of D-617 and [2H3]norverapamil were used as internal standards. The analytes were extracted automatically from plasma and intestinal perfusate using end-capped CN- and C2 solid-phase extraction cartridges. Separation of the eight analytes was achieved on a LUNA C8 analytical column (150x2 mm I.D., 5 microm particle size) with 5 mM ammonium acetate-acetonitrile as the mobile phase run with a gradient from 70:30 to 40:60 and run times of 15 min. With the mass spectrometer operated in the selected-ion monitoring mode, the limits of quantification in plasma and intestinal perfusate were 1 pmol/ml for D-620, D-617, D-702, D-703, norverapamil, verapamil, and [2H7]verapamil and 2.5 pmol/ml for D-717 and D-715 using a sample size of 1 ml plasma and intestinal perfusate. The method described was successfully applied to the determination of verapamil, [2H7]verapamil and their metabolites in human plasma and intestinal fluid in pharmacokinetic studies.


Subject(s)
Body Fluids/chemistry , Chromatography, High Pressure Liquid/methods , Deuterium , Mass Spectrometry/methods , Verapamil/analysis , Verapamil/metabolism , Humans , Intestinal Mucosa/metabolism , Quality Control , Sensitivity and Specificity , Verapamil/blood
11.
J Pharmacol Exp Ther ; 293(2): 376-82, 2000 May.
Article in English | MEDLINE | ID: mdl-10773005

ABSTRACT

Verapamil is subject to extensive oxidative metabolism mediated by cytochrome P450 enzymes with less than 5% of an oral dose being excreted unchanged in urine. Furthermore, verapamil is known to be a potent inhibitor of P-glycoprotein function. There is evidence from in vivo investigations that some verapamil metabolites might be actively transported. The aim of the present study was to investigate P-glycoprotein-mediated transport and inhibition properties of verapamil and its metabolites norverapamil, D-620, D-617, and D-703. Polarized transport of these compounds was assessed in P-glycoprotein-expressing Caco-2 and L-MDR1 cells (LLC-PK1 cells stably transfected with human MDR1-P-glycoprotein). Inhibition of P-glycoprotein-mediated transport by these compounds was determined using digoxin as P-glycoprotein substrate. At concentrations of 5 microM, significant differences between basal-to-apical and apical-to-basal apparent permeability coefficients were observed for D-617 and D-620 in all P-glycoprotein-expressing cell monolayers, indicating that both are P-glycoprotein substrates. In contrast, no P-glycoprotein-dependent transport was found for verapamil, norverapamil, and D-703 in Caco-2 cells and for D-703 in L-MDR1 cells. Moreover, verapamil, norverapamil, and D-703 inhibited P-glycoprotein-mediated digoxin transport with IC(50) values of 1.1, 0.3, and 1.6 microM, respectively, whereas D-617 and D-620 did not (at concentrations up to 100 microM). We conclude that verapamil phase I metabolites exhibit different P-glycoprotein substrate and inhibition characteristics, with the N-dealkylated metabolites D-617 and D-620 being P-glycoprotein substrates and norverapamil and D-703 being inhibitors of P-glycoprotein function, which may influence P-glycoprotein-dependent drug disposition and elimination.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Calcium Channel Blockers/metabolism , Nitriles , Verapamil/analogs & derivatives , Verapamil/metabolism , Animals , Biological Transport, Active , Caco-2 Cells , Calcium Channel Blockers/pharmacology , Cardiotonic Agents/metabolism , Cell Line , Digoxin/metabolism , Epithelial Cells/metabolism , Humans , Kinetics , LLC-PK1 Cells , Swine , Verapamil/pharmacology
12.
Proc Natl Acad Sci U S A ; 97(7): 3473-8, 2000 Mar 28.
Article in English | MEDLINE | ID: mdl-10716719

ABSTRACT

To evaluate whether alterations in the multidrug-resistance (MDR)-1 gene correlate with intestinal MDR-1 expression and uptake of orally administered P-glycoprotein (PGP) substrates, we analyzed the MDR-1 sequence in 21 volunteers whose PGP expression and function in the duodenum had been determined by Western blots and quantitative immunohistology (n = 21) or by plasma concentrations after orally administered digoxin (n = 8 + 14). We observed a significant correlation of a polymorphism in exon 26 (C3435T) of MDR-1 with expression levels and function of MDR-1. Individuals homozygous for this polymorphism had significantly lower duodenal MDR-1 expression and the highest digoxin plasma levels. Homozygosity for this variant was observed in 24% of our sample population (n = 188). This polymorphism is expected to affect the absorption and tissue concentrations of numerous other substrates of MDR-1.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Alleles , Polymorphism, Genetic , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Base Sequence , DNA , Exons , Humans , Intestinal Mucosa/metabolism , Male , Sequence Homology, Nucleic Acid
13.
J Clin Invest ; 104(2): 147-53, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10411543

ABSTRACT

Recent data point to the contribution of P-glycoprotein (P-gp) to digoxin elimination. On the basis of clinical observations of patients in whom digoxin levels decreased considerably when treated with rifampin, we hypothesized that concomitant rifampin therapy may affect digoxin disposition in humans by induction of P-gp. We compared single-dose (1 mg oral and 1 mg intravenous) pharmacokinetics of digoxin before and after coadministration of rifampin (600 mg/d for 10 days) in 8 healthy volunteers. Duodenal biopsies were obtained from each volunteer before and after administration of rifampin. The area under the plasma concentration time curve (AUC) of oral digoxin was significantly lower during rifampin treatment; the effect was less pronounced after intravenous administration of digoxin. Renal clearance and half-life of digoxin were not altered by rifampin. Rifampin treatment increased intestinal P-gp content 3.5 +/- 2.1-fold, which correlated with the AUC after oral digoxin but not after intravenous digoxin. P-gp is a determinant of the disposition of digoxin. Concomitant administration of rifampin reduced digoxin plasma concentrations substantially after oral administration but to a lesser extent after intravenous administration. The rifampin-digoxin interaction appears to occur largely at the level of the intestine. Therefore, induction of intestinal P-gp could explain this new type of drug-drug interaction.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Antibiotics, Antitubercular/pharmacology , Aryl Hydrocarbon Hydroxylases , Cardiotonic Agents/pharmacokinetics , Digoxin/pharmacokinetics , Duodenum/metabolism , Rifampin/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Adult , Carrier Proteins/metabolism , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Duodenum/chemistry , Humans , Male , Microfilament Proteins/metabolism , Oxidoreductases, N-Demethylating/metabolism
14.
Clin Pharmacol Ther ; 65(3): 283-90, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10096260

ABSTRACT

PURPOSE: To quantitate the effect of verapamil administered orally, a calcium channel blocker and potent inhibitor of P-glycoprotein on oral pharmacokinetics of the beta1-adrenergic receptor antagonist talinolol, a substrate of P-glycoprotein. SUBJECTS AND METHODS: In a randomized, crossover placebo-controlled study, oral pharmacokinetics of talinolol (50 mg) after concomitant administration of single doses of R-verapamil (120 mg) or placebo were investigated in 9 healthy volunteers. Concentrations of talinolol, verapamil, and its main metabolite norverapamil were measured in serum with HPLC. Concentrations of talinolol were also measured in urine by HPLC. Standard pharmacokinetic parameters were calculated with noncompartmental procedures. RESULTS: The area under the concentration-time curve for talinolol from 0 to 24 hours was significantly decreased after R-verapamil versus placebo (721+/-231 ng x h x mL(-1) versus 945+/-188 ng x h x mL(-1); P < .01). Maximum serum concentration of talinolol was reached significantly earlier after R-verapamil compared with placebo (P < .05). Coadministration of R-verapamil did not affect the renal clearance or half-life of talinolol. Serum pharmacokinetics are paralleled by the results derived from urine concentrations of talinolol. CONCLUSION: This is the first study to show a decreased oral bioavailability of a P-glycoprotein substrate (talinolol) in humans as a result of coadministration of verapamil. This effect is assumed to be caused by changes of the intestinal net absorption of talinolol because its renal clearance remains unaffected by administration of R-verapamil. This unexpected effect of R-verapamil is most likely dose dependent as a result of an interplay between intestinal P-glycoprotein and gut metabolism.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Adrenergic beta-Antagonists/pharmacokinetics , Calcium Channel Blockers/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Mixed Function Oxygenases/metabolism , Propanolamines/pharmacokinetics , Verapamil/pharmacology , Administration, Oral , Adrenergic beta-Antagonists/administration & dosage , Adult , Biological Availability , Calcium Channel Blockers/administration & dosage , Cross-Over Studies , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/drug effects , Humans , Male , Mixed Function Oxygenases/drug effects , Propanolamines/administration & dosage , Reference Values , Verapamil/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...