Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Clin Pharmacol Ther ; 97(6): 650-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25704243

ABSTRACT

Carboxylesterase 1 (CES1) hydrolyzes the prodrug clopidogrel to an inactive carboxylic acid metabolite. We studied the pharmacokinetics and pharmacodynamics of 600 mg oral clopidogrel in healthy white volunteers, including 10 carriers and 12 noncarriers of CES1 c.428G>A (p.Gly143Glu, rs71647871) single nucleotide variation (SNV). Clopidogrel carboxylic acid to clopidogrel area under the plasma concentration-time curve from 0 hours to infinity (AUC0-∞ ) ratio was 53% less in CES1 c.428G>A carriers than in noncarriers (P = 0.009), indicating impaired hydrolysis of clopidogrel. Consequently, the AUC0-∞ of clopidogrel and its active metabolite were 123% (P = 0.004) and 67% (P = 0.009) larger in the c.428G>A carriers than in noncarriers. Consistent with these findings, the average inhibition of P2Y12 -mediated platelet aggregation 0-12 hours after clopidogrel intake was 19 percentage points higher in the c.428G>A carriers than in noncarriers (P = 0.036). In conclusion, the CES1 c.428G>A SNV increases clopidogrel active metabolite concentrations and antiplatelet effects by reducing clopidogrel hydrolysis to inactive metabolites.


Subject(s)
Carboxylic Ester Hydrolases/genetics , Platelet Aggregation Inhibitors/pharmacology , Polymorphism, Single Nucleotide , Ticlopidine/analogs & derivatives , Adult , Clopidogrel , Female , Genotype , Humans , Hydrolysis , Male , Ticlopidine/pharmacokinetics , Ticlopidine/pharmacology
2.
Br J Pharmacol ; 172(11): 2799-813, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25297798

ABSTRACT

BACKGROUND AND PURPOSE: The effects of ketamine in attenuating morphine tolerance have been suggested to result from a pharmacodynamic interaction. We studied whether ketamine might increase brain morphine concentrations in acute coadministration, in morphine tolerance and morphine withdrawal. EXPERIMENTAL APPROACH: Morphine minipumps (6 mg·day(-1) ) induced tolerance during 5 days in Sprague-Dawley rats, after which s.c. ketamine (10 mg·kg(-1) ) was administered. Tail flick, hot plate and rotarod tests were used for behavioural testing. Serum levels and whole tissue brain and liver concentrations of morphine, morphine-3-glucuronide, ketamine and norketamine were measured using HPLC-tandem mass spectrometry. KEY RESULTS: In morphine-naïve rats, ketamine caused no antinociception whereas in morphine-tolerant rats there was significant antinociception (57% maximum possible effect in the tail flick test 90 min after administration) lasting up to 150 min. In the brain of morphine-tolerant ketamine-treated rats, the morphine, ketamine and norketamine concentrations were 2.1-, 1.4- and 3.4-fold, respectively, compared with the rats treated with morphine or ketamine only. In the liver of morphine-tolerant ketamine-treated rats, ketamine concentration was sixfold compared with morphine-naïve rats. After a 2 day morphine withdrawal period, smaller but parallel concentration changes were observed. In acute coadministration, ketamine increased the brain morphine concentration by 20%, but no increase in ketamine concentrations or increased antinociception was observed. CONCLUSIONS AND IMPLICATIONS: The ability of ketamine to induce antinociception in rats made tolerant to morphine may also be due to increased brain concentrations of morphine, ketamine and norketamine. The relevance of these findings needs to be assessed in humans.


Subject(s)
Analgesics, Opioid/pharmacology , Behavior, Animal/drug effects , Brain/metabolism , Drug Tolerance , Ketamine/pharmacology , Morphine/pharmacology , Analgesics/metabolism , Analgesics/pharmacology , Analgesics, Opioid/metabolism , Animals , Chromatography, High Pressure Liquid , Drug Therapy, Combination , Injections, Subcutaneous , Ketamine/analogs & derivatives , Ketamine/metabolism , Liver/metabolism , Morphine/metabolism , Morphine Derivatives/metabolism , Pain/drug therapy , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry
3.
Clin Pharmacol Ther ; 96(4): 498-507, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24971633

ABSTRACT

Cerivastatin and repaglinide are substrates of cytochrome P450 (CYP)2C8, CYP3A4, and organic anion-transporting polypeptide (OATP)1B1. A recent study revealed an increased risk of rhabdomyolysis in patients using cerivastatin with clopidogrel, warranting further studies on clopidogrel interactions. In healthy volunteers, repaglinide area under the concentration-time curve (AUC(0-∞)) was increased 5.1-fold by a 300-mg loading dose of clopidogrel and 3.9-fold by continued administration of 75 mg clopidogrel daily. In vitro, we identified clopidogrel acyl-ß-D-glucuronide as a potent time-dependent inhibitor of CYP2C8. A physiologically based pharmacokinetic model indicated that inactivation of CYP2C8 by clopidogrel acyl-ß-D-glucuronide leads to uninterrupted 60-85% inhibition of CYP2C8 during daily clopidogrel treatment. Computational modeling resulted in docking of clopidogrel acyl-ß-D-glucuronide at the CYP2C8 active site with its thiophene moiety close to heme. The results indicate that clopidogrel is a strong CYP2C8 inhibitor via its acyl-ß-D-glucuronide and imply that glucuronide metabolites should be considered potential inhibitors of CYP enzymes.


Subject(s)
Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Glucuronides/metabolism , Platelet Aggregation Inhibitors/metabolism , Ticlopidine/analogs & derivatives , Aryl Hydrocarbon Hydroxylases/chemistry , Carbamates/pharmacokinetics , Catalytic Domain , Clopidogrel , Computer Simulation , Cytochrome P-450 CYP2C8/chemistry , Cytochrome P-450 CYP3A/chemistry , Drug Interactions , Female , Humans , Hypoglycemic Agents/pharmacokinetics , Male , Metabolic Detoxication, Phase II , Molecular Docking Simulation , Piperidines/pharmacokinetics , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/pharmacology , Ticlopidine/chemistry , Ticlopidine/metabolism , Ticlopidine/pharmacology , Time Factors
4.
Clin Pharmacol Ther ; 95(3): 307-13, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24067745

ABSTRACT

Cytochrome P450 (CYP) enzymes, including CYP2C19 and CYP3A4, participate in the bioactivation of clopidogrel. Grapefruit juice constituents potently inactivate intestinal CYP3A4 and have been shown to inhibit CYP2C19 as well. In a randomized crossover study, 14 healthy volunteers ingested 200 ml of grapefruit juice or water three times daily for 3 days. On day 3, they ingested a single 600-mg dose of clopidogrel. Grapefruit juice reduced the peak plasma concentration (Cmax) of the active metabolite of clopidogrel to 13% of the control (range 11-17%, P < 0.001) and the area under the plasma concentration-time curve from 0 to 3 h to 14% (range 12-17%, P < 0.001) of the control, but it had no significant effect on the parent clopidogrel. Moreover, grapefruit juice markedly decreased the platelet-inhibitory effect of clopidogrel, as assessed with the VerifyNow P2Y12 test in two of the participants. In conclusion, concomitant use of grapefruit juice may impair the efficacy of clopidogrel. Therefore, the use of grapefruit juice is best avoided during clopidogrel therapy.


Subject(s)
Beverages/adverse effects , Biotransformation/drug effects , Citrus paradisi/adverse effects , Food-Drug Interactions , Platelet Aggregation Inhibitors/metabolism , Ticlopidine/analogs & derivatives , Adult , Area Under Curve , Clopidogrel , Confidence Intervals , Cross-Over Studies , Female , Genotype , Half-Life , Humans , Male , Platelet Function Tests , Receptors, Purinergic P2Y12/genetics , Ticlopidine/metabolism , Young Adult
5.
Eur J Pain ; 18(3): 386-95, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23900882

ABSTRACT

BACKGROUND: Spironolactone, a commonly used mineralocorticoid receptor antagonist, has been reported to potentiate the effect of morphine in the rat. The aim of this study was to investigate the effects of spironolactone on morphine antinociception and tissue distribution. METHODS: The effects of spironolactone on acute morphine-induced antinociception, induction of morphine tolerance and established morphine tolerance were studied with tail-flick and hot plate tests in male Sprague-Dawley rats. Serum, brain, and liver morphine and its metabolite concentrations were quantified using high-pressure liquid chromatography-tandem mass spectrometry. Spironolactone was also administered with the peripherally acting, P-glycoprotein (P-gp) substrate loperamide to test whether spironolactone allows loperamide to pass the blood-brain barrier. RESULTS: Spironolactone (50 mg/kg, i.p.) had no antinociceptive effects of its own, but it enhanced the antinociceptive effect of morphine in both thermal tests. Two doses of spironolactone enhanced the maximum possible effect (MPE) from 19.5% to 100% in the hot plate test 90 min after administration of 4 mg/kg morphine. Morphine concentrations in the brain were increased fourfold at 90 min by spironolactone. Spironolactone did not inhibit the formation of morphine-3-glucuronide. Acute spironolactone restored morphine antinociception in morphine-tolerant rats but did not inhibit the development of tolerance. The peripherally restricted opioid, loperamide (10 mg/kg), had no antinociceptive effects when administered alone, but co-administration with spironolactone produced a 40% MPE in the hot plate test. CONCLUSIONS: Spironolactone has no antinociceptive effects in thermal models of pain, but it enhances the antinociceptive effects of morphine mainly by increasing morphine central nervous system concentrations, probably by inhibiting P-gp.


Subject(s)
Analgesics/therapeutic use , Mineralocorticoid Receptor Antagonists/therapeutic use , Morphine/therapeutic use , Pain/drug therapy , Spironolactone/therapeutic use , Analgesics/pharmacokinetics , Animals , Drug Interactions , Male , Morphine/pharmacokinetics , Pain Measurement , Rats , Rats, Sprague-Dawley , Tissue Distribution
6.
Clin Pharmacol Ther ; 92(1): 68-71, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22588607

ABSTRACT

Bioactivation of the antiviral agent oseltamivir to active oseltamivir carboxylate is catalyzed by carboxylesterase 1 (CES1). After the screening of 860 healthy Finnish volunteers for the CES1 c.428G>A (p.Gly143Glu, rs121912777) polymorphism, a pharmacokinetic study with 75 mg oseltamivir was carried out in c.428G>A carriers and noncarriers. Heterozygous c.428GA carriers (n = 9) had 18% larger values of oseltamivir area under the plasma concentration-time curve from 0 h to infinity (AUC(0-∞)) (P = 0.025) and 23% smaller carboxylate-to-oseltamivir AUC(0-∞) ratio (P = 0.006) than noncarriers (n = 12). This shows that the CES1 c.428G>A polymorphism impairs oseltamivir bioactivation in humans.


Subject(s)
Carboxylic Ester Hydrolases , Influenza A virus/drug effects , Influenza B virus/drug effects , Influenza, Human/drug therapy , Oseltamivir , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Area Under Curve , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Dose-Response Relationship, Drug , Drug Monitoring , Female , Genotype , Humans , Male , Oseltamivir/administration & dosage , Oseltamivir/analogs & derivatives , Oseltamivir/pharmacokinetics , Polymorphism, Genetic , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics
7.
Scand J Pain ; 1(1): 24-33, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-29913934

ABSTRACT

Oxycodone is a strong opioid and it is increasingly used in the management of acute and chronic pain. The pharmacodynamic effects of oxycodone are mainly mediated by the µ-opioid receptor. However, its affinity for the µ-opioid receptor is significantly lower compared with that of morphine and it has been suggested that active metabolites may play a role in oxycodone analgesia. Oxycodone is mainly metabolized by hepatic cytochrome (CYP) enzymes 2D6 and 3A4. Oxycodone is metabolized to oxymorphone, a potent µ-opioid receptor agonist by CYP2D6. However, CYP3A4 is quantitatively a more important metabolic pathway. Chronic pain patients often use multiple medications. Therefore it is important to understand how blocking or inducing these metabolic pathways may affect oxycodone induced analgesia. The aim of this study was to find out whether blocking CYP2D6 would decrease oxycodone induced analgesia in chronic pain patients. The effects of the antidepressant paroxetine, a potent inhibitor of CYP2D6, on the analgesic effects and pharmacokinetics of oral oxycodone were studied in 20 chronic pain patients using a randomized, double-blind, placebo-controlled cross-over study design. Pain intensity and rescue analgesics were recorded daily, and the pharmacokinetics and pharmacodynamics of oxycodone were studied on the 7th day of concomitant paroxetine (20 mg/day) or placebo administration. The patients were genotyped for CYP2D6, 3A4, 3A5 and ABCB1. Paroxetine had significant effects on the metabolism of oxycodone but it had no statistically significant effect on oxycodone analgesia or use of morphine for rescue analgesia. Paroxetine increased the dose-adjusted mean AUC0-12h of oxycodone by 19% (-23 to 113%; P = 0.003), and that of noroxycodone by 100% (5-280%; P < 0.0001) but decreased the AUC0-12 h of oxymorphone by 67% (-100 to -22%; P < 0.0001) and that of noroxymorphone by 68% (-100 to -16%; P < 0.0001). Adverse effects were also recorded in a pain diary for both 7-day periods (placebo/paroxetine). The most common adverse effects were drowsiness and nausea/vomiting. One patient out of four reported dizziness and headache during paroxetine co-administration, whereas no patient reported these during placebo administration (P = 0.0471) indicating that these adverse effects were due to paroxetine. No statistically significant associations of the CYP2D6 or CYP3A4/5 genotype of the patients and the pharmacokinetics of oxycodone or its metabolites, extent of paroxetine-oxycodone interaction, or analgesic effects were observed probably due to the limited number of patients studied. The results of this study strongly suggest that CYP2D6 inhibition does not significantly change oxycodone analgesia in chronic pain patients and that the analgesic activity of oxycodone is mainly due to the parent compound and that metabolites, e.g. oxymorphone, play an insignificant role. The clinical implication of these results is that induction of the metabolism of oxycodone may lead to inadequate analgesia while increased drug effects can be expected after addition of potent CYP3A4/5 inhibitors particularly if combined with CYP2D6 inhibitors or when administered to poor metabolizers of CYP2D6.

8.
J Environ Manage ; 91(4): 984-94, 2010.
Article in English | MEDLINE | ID: mdl-20036782

ABSTRACT

The implementation of the European Union Water Framework Directive (WFD) requires nationally generalizable estimates of the benefits of protecting inland and coastal waters. As an alternative to benefit transfers and meta-analyses, we utilize national recreation inventory data combined with water quality data to model recreation participation and estimate the benefits of water quality improvements. Using hurdle models, we analyze the association of water clarity in individuals' home municipalities with the three most common water recreation activities--swimming, fishing and boating. The results show no effect on boating, but improved water clarity would increase the frequency of close-to-home swimming and fishing, as well as the number of fishers. Furthermore, to value the potential benefits of the WFD, we estimate the consumer surplus of a water recreation day using a travel cost approach. A water policy scenario with a 1-m improvement in water clarity for both inland and coastal waters indicates that the consumer surplus would increase 6% for swimmers and 15% for fishers. In contrast to previously estimated abatement costs to improve water quality, net benefits could turn out to be positive. Our study is a promising example of applying existing national recreation inventory data to estimate the benefits of water quality improvements for the purposes of the WFD.


Subject(s)
Behavior , Conservation of Natural Resources , Environment , Recreation , Travel , Water/standards , Conservation of Natural Resources/economics , Cost-Benefit Analysis , Finland , Fresh Water , Humans , Models, Econometric , Recreation/economics , Seawater , Travel/economics , Travel/statistics & numerical data
9.
Acta Anaesthesiol Scand ; 52(10): 1319-26, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19025521

ABSTRACT

BACKGROUND: Individual variation in opioid response is considerable, partly due to pharmacokinetic factors. Transporter proteins are becoming increasingly interesting also in the pharmacokinetics of opioids. The efflux transporter P-glycoprotein can affect gastrointestinal absorption and tissue distribution, particularly brain access of many opioids. The aim of this study was to evaluate whether itraconazole, which is a potent inhibitor of P-glycoprotein and CYP3A4, would change the pharmacokinetics or the pharmacodynamics of oral morphine. METHODS: Twelve healthy male volunteers ingested, in a randomized crossover study, once daily 200 mg itraconazole or placebo for 4 days. On day 4, 1 h after the last pre-treatment dose, the subjects ingested 0.3 mg/kg morphine. Blood samples for the determination of plasma morphine, morphine-3-glucuronide (M3G), morphine-6-glucuronide (M6G) and itraconazole concentrations were drawn up to 48 h after morphine ingestion. Pharmacodynamic effects were evaluated using a questionnaire, visual analogue scales, a reaction time test, the Digit Symbol Substitution Test and the Critical Flicker Fusion Test. RESULTS: Itraconazole increased the mean area under the plasma concentration-time curve [AUC (0-9)] of morphine by 29% (P=0.002), its AUC (0-48) by 22% (P=0.013) and its peak plasma concentration by 28% (P=0.035). Itraconazole did not significantly affect the pharmacokinetic variables of M3G or M6G or the pharmacodynamic effects of morphine. CONCLUSIONS: Itraconazole moderately increases plasma concentrations of oral morphine, probably by enhancing its absorption by inhibiting intestinal wall P-glycoprotein. A possible improvement of morphine penetration to the brain could not be observed.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Analgesics, Opioid , Antifungal Agents/pharmacology , Itraconazole/pharmacology , Morphine , Administration, Oral , Adult , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/blood , Area Under Curve , Central Nervous System Stimulants/blood , Cross-Over Studies , Double-Blind Method , Humans , Male , Morphine/administration & dosage , Morphine/blood , Morphine Derivatives/blood , Oxygen/blood , Pain Measurement , Research Design
10.
Clin Pharmacol Ther ; 84(3): 403-11, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18388877

ABSTRACT

Repaglinide is metabolized by cytochrome P450 (CYP) 2C8 and 3A4. Gemfibrozil has the effect of increasing the area under the concentration-time curve (AUC) of repaglinide eightfold. We studied the effect of dosing interval on the extent of the gemfibrozil-repaglinide interaction. In a randomized five-phase crossover study, 10 healthy volunteers ingested 0.25 mg repaglinide, with or without gemfibrozil pretreatment. Plasma repaglinide, gemfibrozil, their metabolites, and blood glucose were measured. When the last dose of 600 mg gemfibrozil was ingested simultaneously with repaglinide, or 3, 6, or 12 h before, it increased the AUC(0-infinity) of repaglinide 7.0-, 6.5-, 6.2- and 5.0-fold, respectively (P < 0.001). The peak repaglinide concentration increased approximately twofold (P < 0.001), and the half-life was prolonged from 1.2 h to 2-3 h (P < 0.001) during all the gemfibrozil phases. The drug interaction effects persisted at least 12 h after gemfibrozil was administered, although plasma gemfibrozil and gemfibrozil 1-O-beta-glucuronide concentrations were only 5 and 10% of their peak values, respectively. The long-lasting interaction is likely caused by mechanism-based inhibition of CYP2C8 by gemfibrozil glucuronide.


Subject(s)
Blood Glucose/drug effects , Carbamates/pharmacokinetics , Gemfibrozil/pharmacology , Hypoglycemic Agents/pharmacokinetics , Hypolipidemic Agents/pharmacology , Piperidines/pharmacokinetics , Adult , Area Under Curve , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/genetics , Cytochrome P-450 CYP2C8 , Drug Interactions , Female , Gemfibrozil/analogs & derivatives , Gemfibrozil/blood , Gemfibrozil/pharmacokinetics , Glucuronates/blood , Glucuronates/pharmacokinetics , Half-Life , Humans , Hypolipidemic Agents/blood , Hypolipidemic Agents/pharmacokinetics , Male
11.
Clin Pharmacol Ther ; 84(4): 462-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19238650

ABSTRACT

We compared the pharmacokinetics of 10 mg oral oxycodone in four groups of 10 patients each, aged 20-40, 60-70, 70-80, and 80-90 years. Patients aged 70-80 and 80-90 years had 50-80% higher mean exposure to oxycodone (P < 0.05) and a twofold higher plasma oxycodone concentration (P < 0.05) than the young adults 12 h after ingestion of the drug. Because oxycodone pharmacokinetics depend to a great extent on the age of the subject, it is important to titrate the analgesic dose individually, particularly in the elderly.


Subject(s)
Analgesics, Opioid/blood , Oxycodone/blood , Administration, Oral , Adult , Age Factors , Aged , Aged, 80 and over , Analgesics, Opioid/administration & dosage , Area Under Curve , Female , Humans , Male , Middle Aged , Oxycodone/administration & dosage
12.
Clin Pharmacol Ther ; 81(5): 679-84, 2007 May.
Article in English | MEDLINE | ID: mdl-17301736

ABSTRACT

The aim of this study was to determine the influence of amiodarone on the pharmacokinetics of simvastatin and pravastatin in humans. This was a prospective, crossover, randomized, open-label study performed in 12 healthy volunteers comparing the pharmacokinetics of a single oral dose of simvastatin (40 mg) or pravastatin (40 mg) taken alone and after 3 days of amiodarone (400 mg/day). Amiodarone increased simvastatin acid AUC (area under the plasma concentration-time curve)0-24 h, peak plasma concentration (Cmax), and t1/2 by 73% (P=0.02), 100% (P=0.02), and 48% (P=0.06), respectively, whereas it did not significantly alter pravastatin pharmacokinetics. Point estimates and 90% confidence intervals for simvastatin acid, simvastatin lactone, and pravastatin AUC0-24 h were 154% (109-216%), 155% (109-227%), and 86% (63-118%), respectively. If amiodarone and a statin have to be simultaneously prescribed, pravastatin should be preferred to simvastatin in order to avoid a drug interaction.


Subject(s)
Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Pravastatin/pharmacokinetics , Simvastatin/pharmacokinetics , Adult , Area Under Curve , Biotransformation , Cross-Over Studies , Drug Interactions , Female , Humans , Liver-Specific Organic Anion Transporter 1 , Male , Organic Anion Transporters/biosynthesis , Organic Anion Transporters/genetics , Polymorphism, Genetic/genetics , Prospective Studies , Tissue Distribution
13.
Aliment Pharmacol Ther ; 24(11-12): 1545-52, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17206943

ABSTRACT

BACKGROUND/AIM: To evaluate the safety of budesonide in primary biliary cirrhosis. METHODS: 77 primary biliary cirrhosis patients, with stages I-III at entry, were randomized to use either budesonide 6 mg and ursodeoxycholic acid 15 mg/kg (group A), or ursodeoxycholic acid alone (group B) daily for 3 years. In 22 patients, budesonide pharmacokinetics was determined after 3 years. Bone mass density was measured in 62 patients at baseline and 3 years; in 57 patients also liver biopsies were performed. RESULTS: At 3 years, no significant differences in the pharmacokinetics of budesonide were found between the patients with stages 0-I, II and III primary biliary cirrhosis. In group A, bone mass density in femoral neck and lumbar spine were decreased by 3.6% (P = 0.0002) and 2.8% (P = 0.003) from the baseline. In group B, the corresponding decreases were 1.9% (P = 0.029) and 0.7% (P = 0.25), but the differences between the groups were not statistically significant (P = 0.16 for femoral neck and P = 0.08 for lumbar spine). CONCLUSIONS: The plasma concentrations of budesonide do not significantly differ within stages I-III primary biliary cirrhosis patients. The combination of budesonide and ursodeoxycholic acid may decrease bone mass density in the femoral neck and lumbar spine in some primary biliary cirrhosis patients, and bone mass density is recommended to be monitored during budesonide therapy.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Bone Density/drug effects , Budesonide/adverse effects , Liver Cirrhosis, Biliary/drug therapy , Anti-Inflammatory Agents/pharmacokinetics , Budesonide/pharmacokinetics , Female , Humans , Male , Osteoporosis/prevention & control , Treatment Outcome
14.
Eur J Clin Pharmacol ; 59(12): 879-82, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14691614

ABSTRACT

OBJECTIVE: The aim of this study was to examine the effect of carbamazepine on the pharmacokinetics of orally administered simvastatin in healthy volunteers. METHODS: In a randomised, two-phase crossover study and a wash out of 2 weeks, 12 healthy volunteers took carbamazepine for 14 days (600 mg daily except 200 mg daily for the first 2 days) or no drug. On day 15, each subject ingested 80 mg simvastatin. Serum concentrations of simvastatin and its active metabolite simvastatin acid were measured up to 24 h. RESULTS: Carbamazepine decreased the mean total area under the serum concentration-time curve of simvastatin and simvastatin acid by 75% ( P<0.001) and 82% ( P<0.001), respectively. The mean peak concentrations of both simvastatin and simvastatin acid were reduced by 68% ( P<0.01), and half-life of simvastatin acid was shortened from 5.9+/-0.3 h to 3.7+/-0.5 h ( P<0.01) by carbamazepine. CONCLUSION: Carbamazepine greatly reduces the serum concentrations of simvastatin and simvastatin acid, probably by inducing their metabolism. Concomitant administration of carbamazepine and simvastatin should be avoided or the dose of simvastatin should be considerably increased.


Subject(s)
Anticonvulsants/pharmacology , Carbamazepine/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Simvastatin/analogs & derivatives , Simvastatin/blood , Simvastatin/pharmacokinetics , Adolescent , Adult , Area Under Curve , Cross-Over Studies , Drug Interactions , Half-Life , Humans , Male
15.
Diabetologia ; 46(10): 1319-23, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12898007

ABSTRACT

AIMS/HYPOTHESIS: Our aim was to investigate possible interaction between gemfibrozil and rosiglitazone, a thiazolidinedione antidiabetic drug. METHODS: In a randomised crossover study with two phases, 10 healthy volunteers took 600 mg gemfibrozil or placebo orally twice daily for 4 days. On day 3, they ingested a single 4 mg dose of rosiglitazone. Plasma rosiglitazone and its N-desmethyl metabolite concentrations were measured for up to 48 h. RESULTS: Gemfibrozil raised the mean area under the plasma rosiglitazone concentration-time curve (AUC) 2.3-fold (range 1.5- to 2.8-fold; p=0.00002) and prolonged the elimination half-life (t(1/2)) of rosiglitazone from 3.6 to 7.6 h ( p=0.000002). The peak plasma rosiglitazone concentration (C(max)) was increased only 1.2-fold (range 0.9- to 1.6-fold; p=0.01) by gemfibrozil, but gemfibrozil raised the plasma rosiglitazone concentration measured 24 h after dosing (C(24)) 9.8-fold (range, 4.5- to 33.6-fold; p=0.00008). In addition, gemfibrozil prolonged the t(max) of N-desmethylrosiglitazone from 7 to 12 h and reduced the N-desmethylrosiglitazone/rosiglitazone AUC(0-48) ratio by 38% ( p<0.01). CONCLUSIONS/INTERPRETATION: Gemfibrozil raises the plasma concentrations of rosiglitazone probably by inhibiting the CYP2C8-mediated biotransformation of rosiglitazone. Co-administration of gemfibrozil, or another potent inhibitor of CYP2C8, and rosiglitazone could increase the efficacy but also the risk of concentration-dependent adverse effects of rosiglitazone.


Subject(s)
Gemfibrozil/pharmacology , Hypoglycemic Agents/blood , Hypolipidemic Agents/pharmacology , Thiazolidinediones/blood , Adult , Area Under Curve , Cross-Over Studies , Female , Gemfibrozil/blood , Half-Life , Humans , Hypolipidemic Agents/blood , Male , Osmolar Concentration , Rosiglitazone , Time Factors
16.
Diabetologia ; 46(3): 347-51, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12687332

ABSTRACT

AIMS/HYPOTHESIS: Our aim was to investigate possible interactions of gemfibrozil, itraconazole, and their combination with repaglinide. METHODS: In a randomised crossover study, 12 healthy volunteers received twice daily for 3 days either 600 mg gemfibrozil, 100 mg itraconazole (first dose 200 mg), both gemfibrozil and itraconazole, or placebo. On day 3 they ingested a 0.25 mg dose of repaglinide. Plasma drug and blood glucose concentrations were followed for 7 h and serum insulin and C-peptide concentrations for 3 h postdose. RESULTS: Gemfibrozil raised the area under the plasma concentration-time curve (AUC) of repaglinide 8.1-fold (range 5.5- to 15.0-fold; p<0.001) and prolonged its half-life (t(1/2)) from 1.3 to 3.7 h (p<0.001). Although itraconazole alone raised repaglinide AUC only 1.4-fold (1.1- to 1.9-fold; p<0.001), the gemfibrozil-itraconazole combination raised it 19.4-fold (12.9- to 24.7-fold) and prolonged the t(1/2) of repaglinide to 6.1 h (p<0.001). Plasma repaglinide concentration at 7 h was increased 28.6-fold by gemfibrozil and 70.4-fold by the gemfibrozil-itraconazole combination (p<0.001). Gemfibrozil alone and in combination with itraconazole considerably enhanced and prolonged the blood glucose-lowering effect of repaglinide; i.e., repaglinide became a long-acting and stronger antidiabetic. CONCLUSION/INTERPRETATION: Clinicians should be aware of this previously unrecognised and potentially hazardous interaction between gemfibrozil and repaglinide. Concomitant use of gemfibrozil and repaglinide is best avoided. If the combination is considered necessary, repaglinide dosage should be greatly reduced and blood glucose concentrations carefully monitored.


Subject(s)
Antifungal Agents/adverse effects , Carbamates/pharmacology , Carbamates/pharmacokinetics , Gemfibrozil/adverse effects , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/pharmacokinetics , Hypolipidemic Agents/adverse effects , Itraconazole/adverse effects , Piperidines/pharmacology , Piperidines/pharmacokinetics , Adult , Area Under Curve , Aryl Hydrocarbon Hydroxylases/metabolism , Biotransformation , Blood Glucose/metabolism , C-Peptide/blood , Carbamates/adverse effects , Cross-Over Studies , Cytochrome P-450 CYP2C8 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Female , Half-Life , Humans , Hypoglycemic Agents/adverse effects , Insulin/blood , Male , Piperidines/adverse effects
17.
Clin Pharmacol Ther ; 69(6): 400-6, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11406737

ABSTRACT

OBJECTIVE: To study the effects of rifampin (INN, rifampicin) on the pharmacokinetics and pharmacodynamics of glyburide (INN, glibenclamide) and glipizide, 2 sulfonylurea antidiabetic drugs. METHODS: Two separate, randomized, 2-phase, crossover studies with an identical design were conducted. In each study, 10 healthy volunteers received 600 mg rifampin or placebo once daily for 5 days. On day 6, a single dose of 1.75 mg glyburide (study I) or 2.5 mg glipizide (study II) was administered orally. Plasma glyburide and glipizide and blood glucose concentrations were measured for 12 hours. RESULTS: In study I, rifampin decreased the area under the plasma concentration--time curve [AUC(0-infinity)] of glyburide by 39% (P <.001) and the peak plasma concentration by 22% (P =.01). The elimination half-life of glyburide was shortened from 2.0 to 1.7 hours (P <.05) by rifampin. The blood glucose decremental AUC(0-7) (net area below baseline) and the maximum decrease in the blood glucose concentration were decreased by 44% (P =.05) and 36% (P <.001), respectively, by rifampin. In study II, rifampin decreased the AUC(0-infinity) of glipizide by 22% (P <.05) and shortened its half-life from 3.0 to 1.9 hours (P =.01). No statistically significant differences in the blood glucose concentrations were found between the phases; however, 4 subjects had moderate hypoglycemia during the placebo phase but only 1 subject had moderate hypoglycemia during the rifampin phase. CONCLUSIONS: Rifampin moderately decreased the plasma concentrations and effects of glyburide but had only a slight effect on glipizide. The mechanism underlying the interaction between rifampin and glyburide is probably induction of either CYP2C9 or P-glycoprotein or both. Induction of CYP2C9 would explain the increased systemic elimination of glipizide. It is probable that the blood glucose--lowering effect of glyburide is reduced during concomitant treatment with rifampin. In some patients, the effects of glipizide may also be reduced by rifampin.


Subject(s)
Antibiotics, Antitubercular/pharmacology , Glipizide/pharmacokinetics , Glyburide/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Rifampin/pharmacology , Adult , Area Under Curve , Blood Glucose/metabolism , Female , Glipizide/blood , Glyburide/blood , Half-Life , Humans , Hypoglycemic Agents/blood , Male
18.
Clin Pharmacol Ther ; 69(5): 340-5, 2001 May.
Article in English | MEDLINE | ID: mdl-11372002

ABSTRACT

BACKGROUND: Concomitant use of fibrates with statins has been associated with an increased risk of myopathy, but the underlying mechanism of this adverse reaction remains unclear. Our aim was to study the effects of bezafibrate and gemfibrozil on the pharmacokinetics of lovastatin. METHODS: This was a randomized, double-blind, 3-phase crossover study. Eleven healthy volunteers took 400 mg/day bezafibrate, 1200 mg/day gemfibrozil, or placebo for 3 days. On day 3, each subject ingested a single 40 mg dose of lovastatin. Plasma concentrations of lovastatin, lovastatin acid, gemfibrozil, and bezafibrate were measured up to 24 hours. RESULTS: Gemfibrozil markedly increased the plasma concentrations of lovastatin acid, without affecting those of the parent lovastatin compared with placebo. During the gemfibrozil phase, the mean area under the plasma concentration-time curve from 0 to 24 hours [AUC(0-24)] of lovastatin acid was 280% (range, 131% to 1184%; P < .001) and the peak plasma concentration (Cmax) was 280% (range, 123% to 1042%; P < .05) of the corresponding value during the placebo phase. Bezafibrate had no statistically significant effect on the AUC(0-24) or Cmax of lovastatin or lovastatin acid compared with placebo. CONCLUSIONS: Gemfibrozil markedly increases plasma concentrations of lovastatin acid, but bezafibrate does not. The increased risk of myopathy observed during concomitant treatment with statins and fibrates may be partially of a pharmacokinetic origin. The risk of developing myopathy during concomitant therapy with lovastatin and a fibrate may be smaller with bezafibrate than with gemfibrozil.


Subject(s)
Bezafibrate/pharmacology , Gemfibrozil/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hypolipidemic Agents/pharmacology , Lovastatin/pharmacokinetics , Adult , Area Under Curve , Bezafibrate/blood , Cross-Over Studies , Drug Interactions , Female , Gemfibrozil/blood , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hypolipidemic Agents/blood , Lovastatin/blood , Male
19.
Clin Pharmacol Ther ; 69(4): 194-200, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309547

ABSTRACT

OBJECTIVE: Our objective was to study the effects of fluconazole and fluvoxamine on the pharmacokinetics and pharmacodynamics of glimepiride, a new sulfonylurea antidiabetic drug. METHODS: In this randomized, double-blind, three-phase crossover study, 12 healthy volunteers took 200 mg of fluconazole once daily (400 mg on day 1), 100 mg of fluvoxamine once daily, or placebo once daily for 4 days. On day 4, a single oral dose of 0.5 mg of glimepiride was administered. Plasma glimepiride and blood glucose concentrations were measured up to 12 hours. RESULTS: In the fluconazole phase, the mean total area under the plasma concentration-time curve of glimepiride was 238% (P <.0001) and the peak plasma concentration was 151% (P <.0001) of the respective control value. The mean elimination half-life of glimepiride was prolonged from 2.0 to 3.3 hours (P <.0001) by fluconazole. In the fluvoxamine phase, the mean area under the plasma concentration-time curve of glimepiride was not significantly different from that in the placebo phase. However, the mean peak plasma concentration of glimepiride was 143% (P <.05) of the control and the elimination half-life was prolonged from 2.0 to 2.3 hours (P <.01) by fluvoxamine. Fluconazole and fluvoxamine did not cause statistically significant changes in the effects of glimepiride on blood glucose concentrations. CONCLUSIONS: Fluconazole considerably increased the area under the plasma concentration-time curve of glimepiride and prolonged its elimination half-life. This was probably caused by inhibition of the cytochrome P-450 2C9-mediated biotransformation of glimepiride by fluconazole. Concomitant use of fluconazole with glimepiride may increase the risk of hypoglycemia as much as would a 2- to 3-fold increase in the dose of glimepiride. Fluvoxamine moderately increased the plasma concentrations and slightly prolonged the elimination half-life of glimepiride.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Fluconazole/pharmacology , Fluvoxamine/pharmacology , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/pharmacokinetics , Steroid 16-alpha-Hydroxylase , Sulfonylurea Compounds/pharmacology , Sulfonylurea Compounds/pharmacokinetics , Adult , Area Under Curve , Blood Glucose/analysis , Cross-Over Studies , Cytochrome P-450 Enzyme Inhibitors , Double-Blind Method , Drug Interactions , Female , Fluconazole/pharmacokinetics , Fluvoxamine/pharmacokinetics , Half-Life , Humans , Male , Steroid Hydroxylases/antagonists & inhibitors
20.
Clin Pharmacol Ther ; 68(5): 495-500, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11103752

ABSTRACT

OBJECTIVE: To study the effects of rifampin (INN, rifampicin) on the pharmacokinetics and pharmacodynamics of repaglinide, a new short-acting antidiabetic drug. METHODS: In a randomized, two-phase crossover study, nine healthy volunteers were given a 5-day pretreatment with 600 mg rifampin or matched placebo once daily. On day 6 a single 0.5-mg dose of repaglinide was administered. Plasma repaglinide and blood glucose concentrations were measured up to 7 hours. RESULTS: Rifampin decreased the total area under the concentration-time curve of repaglinide by 57% (P < .001) and the peak plasma repaglinide concentration by 41% (P = .001). The elimination half-life of repaglinide was shortened from 1.5 to 1.1 hours (P < .01). The blood glucose decremental area under the concentration-time curve from 0 to 3 hours was reduced from 0.94 to -0.23 mmol/L x h (P < .05), and the maximum decrease in blood glucose concentration from 1.6 to 1.0 mmol/L (P < .05) by rifampin. CONCLUSIONS: Rifampin considerably decreases the plasma concentrations of repaglinide and also reduces its effects. This interaction is probably caused by induction of the CYP3A4-mediated metabolism of repaglinide. It is probable that the effects of repaglinide are decreased during treatment with rifampin or other potent inducers of CYP3A4, such as carbamazepine, phenytoin, or St John's wort.


Subject(s)
Carbamates/pharmacology , Carbamates/pharmacokinetics , Enzyme Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/pharmacokinetics , Piperidines/pharmacology , Piperidines/pharmacokinetics , Rifampin/pharmacology , Adult , Area Under Curve , Blood Glucose/drug effects , Carbamates/blood , Cross-Over Studies , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Drug Interactions , Female , Half-Life , Humans , Hypoglycemic Agents/blood , Male , Mixed Function Oxygenases/antagonists & inhibitors , Piperidines/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...