Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
J Matern Fetal Neonatal Med ; 28(17): 2015-9, 2015.
Article in English | MEDLINE | ID: mdl-25354289

ABSTRACT

OBJECTIVE: Opioid exposure during pregnancy is a potential risk factor for the developing central nervous system of the fetus. We studied evoked responses in buprenorphine-exposed newborns who displayed neonatal abstinence syndrome (NAS) to elucidate the possible alterations in functioning of the somatosensory and auditory systems. METHODS: We compared somatosensory (SEFs) and auditory evoked magnetic fields (AEFs), recorded with magnetoencephalography (MEG), of 11 prenatally buprenorphine-exposed newborns with those of 12 healthy newborns. Peak latencies, source strength and location of SEFs or AEFs were recorded. RESULTS: AEFs were present in all buprenorphine-exposed newborns without significant differences from those of healthy newborns. In contrast, though no group level differences in SEFs existed, at individual level the response deviated from the typical neonatal morphology in four buprenorphine-exposed newborns. CONCLUSIONS: Although buprenorphine exposure during pregnancy does not seem to cause constant deficiencies in somatosensory or auditory processing, in some newborns the typical development of somatosensory networks may be - at least transiently - disrupted.


Subject(s)
Buprenorphine/adverse effects , Evoked Potentials, Auditory , Evoked Potentials, Somatosensory , Magnetoencephalography , Maternal-Fetal Exchange , Neonatal Abstinence Syndrome/physiopathology , Analgesics, Opioid/adverse effects , Auditory Pathways/drug effects , Auditory Pathways/physiopathology , Female , Gestational Age , Humans , Infant, Newborn , Male , Neonatal Abstinence Syndrome/etiology , Pregnancy , Somatosensory Disorders/diagnosis , Somatosensory Disorders/physiopathology
2.
Eur Arch Paediatr Dent ; 15(3): 197-202, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24132830

ABSTRACT

PURPOSE: To study the oral health and dental neglect of prenatally buprenorphine-exposed 3-year-old children. METHODS: The study consisted of 51 children who as newborns tested positive for buprenorphine in a urine screen. The control group comprised 68 children previously unexposed to narcotics. The dentist examined the children and interviewed their guardians. RESULTS: Buprenorphine-exposed children exhibited significantly more early childhood caries than did the control group. Caries indices, the number of decayed, missing and filled teeth or tooth surfaces and decayed teeth were greater in the buprenorphine-exposed children than the control children (p = 0.004, p = 0.004, p = 0.001, respectively). In the buprenorphine group, more children showed visible plaque (p = 0.003) and fewer children were caries-free (p = 0.009) than in the control group. The control children's teeth were also brushed more often than the buprenorphine-exposed children's teeth (p = 0.001) and the parents were more involved in their children's tooth brushing than were those in the buprenorphine-exposed group (p = 0.035). CONCLUSIONS: More caries and dental neglect were found in buprenorphine-exposed children than in controls. These findings highlight the importance of routine dental appointments, caries screening and preventive care for children in substance-abusing families.


Subject(s)
Buprenorphine/adverse effects , Child Abuse/diagnosis , Narcotics/adverse effects , Opioid-Related Disorders , Oral Health , Prenatal Exposure Delayed Effects , Adult , Child Care , Child, Preschool , DMF Index , Dental Care for Children , Dental Caries/diagnosis , Dental Enamel/abnormalities , Dental Plaque/diagnosis , Educational Status , Female , Humans , Male , Oral Hygiene , Parent-Child Relations , Parents/education , Pregnancy , Smoking , Social Class , Toothbrushing
3.
Clin Pharmacol Ther ; 84(1): 104-10, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18167502

ABSTRACT

We studied the efficacy of propafenone in preventing atrial tachyarrhythmias after cardiac surgery, and the possible relationships between CYP2D6 polymorphism and the efficacy, pharmacokinetics, and tolerability of propafenone. One hundred and sixty patients were randomized (double blind) to receive propafenone (n= 78) or placebo (n= 82) for 1 week after cardiac surgery. The patients who were assigned to the propafenone group received 1 mg/kg infused in 1 h, followed by a continuous infusion at a rate of 4 mg/kg/24 h until the following morning, and subsequently 450 mg/day orally until the sixth postoperative day. Thirty-seven patients completed the trial in the propafenone group and 45 in the placebo group. The frequency of occurrence of atrial tachyarrhythmia was lower in the propafenone group than in the placebo group (29.7% vs. 53.3%, P< 0.05; relative risk, 0.56). Plasma propafenone concentrations were markedly influenced by CYP2D6 genotype-derived phenotype.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/prevention & control , Propafenone/therapeutic use , Tachycardia/prevention & control , Thoracic Surgery , Aged , Anti-Arrhythmia Agents/blood , Atrial Fibrillation/enzymology , Atrial Fibrillation/genetics , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Double-Blind Method , Female , Humans , Male , Middle Aged , Polymorphism, Genetic/genetics , Postoperative Complications/enzymology , Postoperative Complications/prevention & control , Propafenone/blood , Tachycardia/enzymology , Tachycardia/genetics
4.
Neurology ; 69(11): 1152-9, 2007 Sep 11.
Article in English | MEDLINE | ID: mdl-17846414

ABSTRACT

OBJECTIVE: Dysfunction of mitochondrial DNA polymerase gamma (POLG) has been recently recognized as an important cause of inherited neurodegenerative diseases. We have reported dominant and recessive inheritance of parkinsonism, mitochondrial myopathy, and premature amenorrhea in five ethnically distinct families with POLG1 mutations. This prompted us to carry out a detailed analysis of the coding region and intron-exon boundaries of POLG1 in Finnish patients with idiopathic sporadic Parkinson disease (PD) and in nonparkinsonian controls. METHODS: The coding region of POLG1 was analyzed in 140 Finnish patients with PD and their 127 spouses as age- and ethnically matched controls. Further, we analyzed the intragenic CAG-repeat region of POLG1 in 126 additional patients with nonparkinsonian neurologic disorders and in 516 Finnish population controls. RESULTS: We found clustering of rare variants of the POLG1 CAG-repeat, encoding a polyglutamine tract, in Finnish patients with idiopathic PD as compared to their spouses (p = 0.003; OR 3.01, 95% CI 1.35 to 6.71), population controls (p = 0.001; OR 2.45, 95% CI 1.45 to 4.14), and patients with nonparkinsonian neurologic disorders (p = 0.05, OR 1.98, 95% CI 0.97 to 4.05). We found several amino acid substitutions, none of them associating with PD. These included a previously parkinsonism-associated POLG variant Y831C, found in one patient with PD, but also in five controls, suggesting that it is a neutral amino acid polymorphism. CONCLUSIONS: Our results suggest that POLG polyglutamine tract variants should be considered as a predisposing genetic factor in idiopathic sporadic Parkinson disease.


Subject(s)
DNA, Mitochondrial/genetics , DNA-Directed DNA Polymerase/genetics , Genetic Predisposition to Disease/genetics , Parkinson Disease/enzymology , Parkinson Disease/genetics , Polymorphism, Genetic/genetics , Adult , Aged , Aged, 80 and over , DNA Mutational Analysis , DNA Polymerase gamma , DNA, Mitochondrial/biosynthesis , Female , Genetic Markers/genetics , Genetic Testing , Genotype , Humans , Male , Middle Aged , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/physiopathology , Open Reading Frames/genetics , Parkinson Disease/physiopathology , Peptides/genetics , Trinucleotide Repeat Expansion/genetics
5.
Br J Clin Pharmacol ; 52(5): 547-53, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11736863

ABSTRACT

AIMS: To evaluate the potency and specificity of valproic acid as an inhibitor of the activity of different human CYP isoforms in liver microsomes. METHODS: Using pooled human liver microsomes, the effects of valproic acid on seven CYP isoform specific marker reactions were measured: phenacetin O-deethylase (CYP1A2), coumarin 7-hydroxylase (CYP2A6), tolbutamide hydroxylase (CYP2C9), S-mephenytoin 4'-hydroxylase (CYP2C19), dextromethorphan O-demethylase (CYP2D6), chlorzoxazone 6-hydroxylase (CYP2E1) and midazolam 1'-hydroxylase (CYP3A4). RESULTS: Valproic acid competitively inhibited CYP2C9 activity with a Ki value of 600 microM. In addition, valproic acid slightly inhibited CYP2C19 activity (Ki = 8553 microM, mixed inhibition) and CYP3A4 activity (Ki = 7975 microM, competitive inhibition). The inhibition of CYP2A6 activity by valproic acid was time-, concentration- and NADPH-dependent (KI = 9150 microM, Kinact=0.048 min(-1)), consistent with mechanism-based inhibition of CYP2A6. However, minimal inhibition of CYP1A2, CYP2D6 and CYP2E1 activities was observed. CONCLUSIONS: Valproic acid inhibits the activity of CYP2C9 at clinically relevant concentrations in human liver microsomes. Inhibition of CYP2C9 can explain some of the effects of valproic acid on the pharmacokinetics of other drugs, such as phenytoin. Co-administration of high doses of valproic acid with drugs that are primarily metabolized by CYP2C9 may result in significant drug interactions.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/pharmacology , Microsomes, Liver/drug effects , Steroid 16-alpha-Hydroxylase , Valproic Acid/pharmacology , Cytochrome P-450 CYP1A2 , Cytochrome P-450 CYP2A6 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 Enzyme Inhibitors , Dose-Response Relationship, Drug , Humans , Hydroxylation/drug effects , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Microsomes, Liver/enzymology , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/metabolism , Steroid Hydroxylases/antagonists & inhibitors , Steroid Hydroxylases/metabolism , Time Factors , Tolbutamide/metabolism
6.
Eur J Clin Pharmacol ; 57(6-7): 457-60, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11699609

ABSTRACT

OBJECTIVE: To study the effects of methylprednisolone on the pharmacokinetics and pharmacodynamics of triazolam. METHODS: In this three-phase cross-over study, ten healthy subjects received 0.25 mg oral triazolam on three occasions: on day 1 (no pretreatment, control), on day 8 (1 h after a single dose of 32 mg oral methylprednisolone) and on day 18 (after further treatment with 8 mg oral methylprednisolone daily for 9 days). The plasma concentrations of triazolam were determined up to 10 h, and its effects were measured using four psychomotor tests up to 6 h. RESULTS: The single dose of methylprednisolone showed no significant effects on the pharmacokinetics of triazolam. However, the Digit Symbol Substitution Test result was better (P < 0.05) during the single-dose methylprednisolone phase than during the control phase, the other three tests showing no differences between the phases. The multiple-dose treatment with methylprednisolone reduced the mean peak plasma concentration (Cmax) of triazolam by 30% (P < 0.05) but had no significant effects on the time to Cmax (tmax), elimination half-life (t 1/2), area under the plasma concentration-time curve from 0 h to 10 h (AUC(0-10 h)) and AUC(0-infinity) and did not alter the effects of triazolam. CONCLUSION: A single, relatively high dose of methylprednisolone (32 mg) did not affect cytochrome P450 (CYP)3A4 activity, and treatment with 8 mg methylprednisolone daily for 9 days did not result in clinically significant induction of CYP3A4.


Subject(s)
Anti-Anxiety Agents/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Glucocorticoids/pharmacokinetics , Methylprednisolone/pharmacokinetics , Mixed Function Oxygenases/metabolism , Triazolam/pharmacokinetics , Adult , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/blood , Anti-Anxiety Agents/pharmacology , Area Under Curve , Cross-Over Studies , Cytochrome P-450 CYP3A , Female , Glucocorticoids/pharmacology , Humans , Male , Methylprednisolone/pharmacology , Psychomotor Performance/drug effects , Triazolam/administration & dosage , Triazolam/blood , Triazolam/pharmacology
7.
Clin Pharmacol Ther ; 70(5): 439-45, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11719730

ABSTRACT

OBJECTIVE: Our objective was to study the effects of gemfibrozil on the pharmacokinetics and pharmacodynamics of glimepiride, a new sulfonylurea antidiabetic drug and a substrate of cytochrome P4502C9 (CYP2C9). METHODS: In a randomized, 2-phase crossover study, 10 healthy volunteers were treated for 2 days with 600 mg oral gemfibrozil or placebo twice daily. On day 3, they received a single dose of 600 mg gemfibrozil or placebo and 1 hour later a single dose of 0.5 mg glimepiride orally. Plasma glimepiride, serum insulin, and blood glucose concentrations were measured up to 12 hours. RESULTS: Gemfibrozil increased the mean total area under the plasma concentration-time curve of glimepiride by 23% (range, 6%-56%; P <.005). The mean elimination half-life of glimepiride was prolonged from 2.1 to 2.3 hours (P <.05) by gemfibrozil. No statistically significant differences were found in the serum insulin or blood glucose variables between the two phases. CONCLUSIONS: Gemfibrozil modestly increases the plasma concentrations of glimepiride. This may be caused by inhibition of CYP2C9.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Gemfibrozil/pharmacology , Hypoglycemic Agents/pharmacokinetics , Hypolipidemic Agents/pharmacology , Steroid 16-alpha-Hydroxylase , Sulfonylurea Compounds/pharmacokinetics , Adult , Area Under Curve , Cross-Over Studies , Cytochrome P-450 CYP2C9 , Cytochrome P-450 Enzyme System/physiology , Drug Interactions , Female , Humans , Male , Steroid Hydroxylases/physiology , Sulfonylurea Compounds/pharmacology
8.
Br J Clin Pharmacol ; 52(4): 399-407, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11678783

ABSTRACT

AIMS: To determine whether the pharmacokinetics of cisapride and its interaction with grapefruit juice are stereoselective. METHODS: The study was a randomized, two-phase cross over design with a washout period of 2 weeks. Ten healthy volunteers were pretreated with either water or 200 ml double strength grapefruit juice three times a day for 2 days. On the 3rd each subject ingested a single 10 mg dose of rac-cisapride tablet. Double strength grapefruit juice (200 ml) or water was administered during cisapride dosing and 0.5 and 1.5 h thereafter. Blood samples were collected before and for 32 h after cisapride administration. Plasma concentrations of cisapride enantiomers were measured by a chiral h.p.l.c. method. A standard 12-lead ECG was recorded before cisapride administration (baseline) and 2, 5, 8, and 12 h later. RESULTS: This study showed that cisapride pharmacokinetics are stereoselective. In control (water treated) subjects, the mean Cmax (30 +/- 13.6 ng ml-1; P = 0.0008) and AUC(0, infinity) (201 +/- 161 ng ml-1 h; P = 0.029) of (-)-cisapride were significantly higher than the Cmax (10.5 +/- 3.4 ng ml-1) and AUC(0, infinity) (70 +/- 51.5 ng ml-1 h) of (+)-cisapride. There was no marked difference in elimination half-life between (-)-cisapride (4.7 +/- 2.7 h) and (+)-cisapride (4.8 +/- 3 h). Compared with the water treated group, grapefruit juice significantly increased the mean Cmax of (-)-cisapride from 30 +/- 13.6-55.5 +/- 18 ng ml-1 (95% CI on mean difference, -33, -17; P = 0.00005) and of (+)-cisapride from 10.5 +/- 3.4 to 18.4 +/- 6.2 ng ml-1 (95% CI on mean difference, -11.8, -3.9, P = 0.00015). The mean AUC(0, infinity) of (-)-cisapride was increased from 201 +/- 161 to 521.6 +/- 303 ng ml-1 h (95% CI on mean difference, -439, -202; P = 0.0002) and that of (+)-cisapride from 70 +/- 51.5 to 170 +/- 91 ng ml-1 h (95% CI on mean difference, -143, -53; P = 0.0005). The tmax was also significantly increased for both enantiomers (from 1.35 to 2.8 h for (-)-cisapride and from 1.75 to 2.9 h for (+)-cisapride in the control and grapefruit juice group, respectively; P < 0.05). The t(1/2) of (-)-cisapride was significantly increased by grapefruit juice, while this change did not reach significant level for (+)-cisapride. The proportion of pharmacokinetic changes brought about by grapefruit juice was similar for both enantiomers, suggesting non-stereoselective interaction. We found no significant difference in mean QTc intervals between the water and grapefruit juice treated groups. CONCLUSIONS: The pharmacokinetics of cisapride is stereoselective. Grapefruit juice elevates plasma concentrations of both (-)- and (+)-cisapride, probably through inhibition of CYP3A in the intestine. At present, there are no data on whether the enantiomers exhibit stereoselective pharmacodynamic actions. If they do, determination of plasma concentrations of the individual enantiomers as opposed to those of racemic cisapride may better predict the degree of drug interaction, cardiac safety and prokinetic efficacy of cisapride.


Subject(s)
Beverages , Cisapride/pharmacokinetics , Citrus , Gastrointestinal Agents/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Cisapride/administration & dosage , Dose-Response Relationship, Drug , Drug Interactions , Electrocardiography/drug effects , Gastrointestinal Agents/administration & dosage , Half-Life , Humans , Metabolic Clearance Rate , Stereoisomerism
9.
Drug Metab Dispos ; 29(11): 1359-61, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11602509

ABSTRACT

The in vitro inhibitory effects of gemfibrozil on cytochrome P450 (CYP) 1A2 (phenacetin O-deethylation), CYP2A6 (coumarin 7-hydroxylation), CYP2C9 (tolbutamide hydroxylation), CYP2C19 (S-mephenytoin 4'-hydroxylation), CYP2D6 (dextromethorphan O-deethylation), CYP2E1 (chlorzoxazone 6-hydroxylation), and CYP3A4 (midazolam 1'-hydroxylation) activities were examined using pooled human liver microsomes. The in vivo drug interactions of gemfibrozil were predicted in vitro using the [I]/([I] + K(i)) values. Gemfibrozil strongly and competitively inhibited CYP2C9 activity, with a K(i) (IC(50)) value of 5.8 (9.6) microM. In addition, gemfibrozil exhibited somewhat smaller inhibitory effects on CYP2C19 and CYP1A2 activities, with K(i) (IC(50)) values of 24 (47) microM and 82 (136) microM, respectively. With concentrations up to 250 microM, gemfibrozil showed no appreciable effect on CYP2A6, CYP2D6, CYP2E1, and CYP3A4 activities. Based on [I]/([I] + K(i)) values calculated using peak total (or unbound) plasma concentration of gemfibrozil, 96% (56%), 86% (24%), and 64% (8%) inhibition of the clearance of CYP2C9, CYP2C19, and CYP1A2 substrates could be expected, respectively. In conclusion, gemfibrozil inhibits the activity of CYP2C9 at clinically relevant concentrations, and this is the likely mechanism by which gemfibrozil interacts with CYP2C9 substrate drugs, such as warfarin and glyburide. Gemfibrozil may also impair clearance of CYP2C19 and CYP1A2 substrates, but inhibition of other CYP isoforms is unlikely.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Gemfibrozil/pharmacology , Hypolipidemic Agents/pharmacology , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/antagonists & inhibitors , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP1A2 Inhibitors , Cytochrome P-450 CYP2A6 , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Female , Humans , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Mixed Function Oxygenases/metabolism , Steroid Hydroxylases/metabolism
10.
Clin Pharmacol Ther ; 70(3): 255-60, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11557913

ABSTRACT

OBJECTIVE: Our objective was to study the effect of activated charcoal on the absorption of sustained-release drugs ingested 1 hour earlier and to examine whether whole-bowel irrigation affects the efficacy of charcoal. METHODS: In this randomized, 3-phase crossover study, 9 healthy subjects received, at the same time, 200 mg carbamazepine, 200 mg theophylline, and 120 mg verapamil. All drugs were given as sustained-release tablets. One hour after taking the tablets, the subjects were assigned to one of the following treatments: 25 g activated charcoal as a suspension, 25 g activated charcoal as a suspension followed by whole-bowel irrigation with polyethylene glycol (PEG) electrolyte lavage solution, or 200 mL water (control). The absorption of the drugs was characterized by using the area under the plasma drug concentration-time curve from time zero to 24 hours [AUC(0-24)], peak plasma concentration (C(max)), C(max) minus the plasma concentration at 1 hour (C(Delta)), and time to peak (t(max)). RESULTS: Activated charcoal alone given 1 hour after drug intake significantly (P <.001) reduced the absorption [AUC(0-24)] of all 3 drugs (by 62%-75%). Also the C(max) and C(Delta) values of these drugs were significantly reduced by charcoal alone. Whole-bowel irrigation did not increase significantly the effect of charcoal on any absorption parameters of the 3 drugs studied. On the contrary, whole-bowel irrigation significantly (P <.01) decreased the efficacy of charcoal with respect to carbamazepine. CONCLUSIONS: Activated charcoal alone given 1 hour after intake of sustained-release drugs was effective in preventing the absorption of all 3 drugs studied. Whole-bowel irrigation may even decrease the efficacy of charcoal if the drug is well adsorbable onto charcoal. However, our study was performed with therapeutic drug doses only. In overdoses their possible effects on gastrointestinal motility may modify the efficacy of decontamination methods.


Subject(s)
Antidotes/pharmacology , Charcoal/pharmacology , Delayed-Action Preparations/pharmacokinetics , Intestinal Absorption/drug effects , Therapeutic Irrigation , Adult , Anticonvulsants/administration & dosage , Anticonvulsants/pharmacokinetics , Area Under Curve , Bronchodilator Agents/administration & dosage , Bronchodilator Agents/pharmacokinetics , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/pharmacokinetics , Carbamazepine/administration & dosage , Carbamazepine/pharmacokinetics , Cross-Over Studies , Double-Blind Method , Female , Humans , Male , Theophylline/administration & dosage , Theophylline/pharmacokinetics , Verapamil/administration & dosage , Verapamil/pharmacokinetics
11.
Clin Pharmacol Ther ; 70(1): 58-65, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11452245

ABSTRACT

OBJECTIVE: Our objective was to study the effects of the macrolide antibiotic clarithromycin on the pharmacokinetics and pharmacodynamics of repaglinide, a novel short-acting antidiabetic drug. METHODS: In a randomized, double-blind, 2-phase crossover study, 9 healthy volunteers were treated for 4 days with 250 mg oral clarithromycin or placebo twice daily. On day 5 they received a single dose of 250 mg clarithromycin or placebo, and 1 hour later a single dose of 0.25 mg repaglinide was given orally. Plasma repaglinide, serum insulin, and blood glucose concentrations were measured up to 7 hours. RESULTS: Clarithromycin increased the mean total area under the concentration-time curve of repaglinide by 40% (P <.0001) and the peak plasma concentration by 67% (P <.005) compared with placebo. The mean elimination half-life of repaglinide was prolonged from 1.4 to 1.7 hours (P <.05) by clarithromycin. Clarithromycin increased the mean incremental area under the concentration-time curve from 0 to 3 hours of serum insulin by 51% (P <.05) and the maximum increase in the serum insulin concentration by 61% (P <.01) compared with placebo. No statistically significant differences were found in the blood glucose concentrations between the placebo and clarithromycin phases. CONCLUSIONS: Even low doses of the cytochrome P4503A4 (CYP3A4) inhibitor clarithromycin increase the plasma concentrations and effects of repaglinide. Concomitant use of clarithromycin or other potent inhibitors of CYP3A4 with repaglinide may enhance its blood glucose-lowering effect and increase the risk of hypoglycemia.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carbamates/blood , Carbamates/pharmacology , Clarithromycin/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Hypoglycemic Agents/blood , Hypoglycemic Agents/pharmacology , Mixed Function Oxygenases/antagonists & inhibitors , Piperidines/blood , Piperidines/pharmacology , Adult , Analysis of Variance , Anti-Bacterial Agents/administration & dosage , Blood Glucose/metabolism , Carbamates/administration & dosage , Carbamates/pharmacokinetics , Clarithromycin/administration & dosage , Cross-Over Studies , Cytochrome P-450 CYP3A , Double-Blind Method , Drug Administration Schedule , Female , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacokinetics , Insulin/blood , Male , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Reference Values , Time Factors
12.
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
13.
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
14.
Eur J Clin Pharmacol ; 57(1): 37-42, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11372588

ABSTRACT

OBJECTIVE: To characterise the effects of itraconazole, a potent inhibitor of CYP3A4, on the pharmacokinetics of selegiline in healthy volunteers. METHODS: In this randomised, placebo-controlled crossover study with two phases, 12 healthy volunteers took either 200 mg itraconazole or matched placebo once daily for 4 days. On day 4, a single 10-mg oral dose of selegiline hydrochloride was administered. Serum concentrations of selegiline and its primary metabolites desmethylselegiline and l-methamphetamine were determined up to 32 h. A caffeine test was performed on day 3 of both phases, by measuring the plasma paraxanthine/caffeine concentration ratio 6 h after caffeine intake, to examine the role of CYP1A2 in selegiline pharmacokinetics. In addition, the effects of itraconazole on the metabolism of selegiline in vitro were characterised by using human liver microsomes. RESULTS: Itraconazole had no significant effects on the pharmacokinetic variables of selegiline, desmethylselegiline or l-methamphetamine, with the exception that the AUC of desmethylselegiline was increased by about 10% (P < 0.05). There was a significant correlation between the AUC(desmethylselegiline)/AUC(selegiline) ratio and the paraxanthine/caffeine ratio (r = 0.41; P < 0.05), suggesting involvement of CYP1A2 in the formation of desmethylselegiline. In experiments with human liver microsomes, itraconazole had no inhibitory effect on the formation of either desmethylselegiline or l-methamphetamine from selegiline. CONCLUSIONS: The pharmacokinetics of selegiline in healthy volunteers were unaffected by the potent CYP3A4 inhibitor itraconazole. In addition, itraconazole showed no inhibitory effect on the biotransformation of selegiline to desmethylselegiline and l-methamphetamine by human liver microsomes. These findings suggest that selegiline is not susceptible to interaction with CYP3A4 inhibitors.


Subject(s)
Antifungal Agents/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Itraconazole/pharmacology , Microsomes, Liver/enzymology , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/metabolism , Selegiline/pharmacokinetics , Adult , Amphetamines/blood , Amphetamines/pharmacokinetics , Antifungal Agents/blood , Area Under Curve , Central Nervous System Stimulants/blood , Central Nervous System Stimulants/pharmacokinetics , Confidence Intervals , Cross-Over Studies , Cytochrome P-450 CYP3A , Drug Interactions/physiology , Female , Humans , Itraconazole/blood , Male , Methamphetamine/blood , Methamphetamine/pharmacokinetics , Selegiline/blood , Statistics, Nonparametric
15.
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
16.
Ther Drug Monit ; 22(6): 749-52, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11128245

ABSTRACT

The aim of this study was to assess the reliability of patient history in the identification of the drugs taken by patients who have an acute drug overdose. To this end, a prospective study involving 51 cases of acute, deliberate drug poisoning was carried out (patients with ethanol as the only apparent cause of intoxication were excluded). Information based on interviews with the patients and their companions or on circumstantial evidence (e.g., drug containers found) was compared with the results from drug analyses of various body fluids. The information obtained on admission was completely in accordance with the laboratory findings in only 27% of the cases. Minor discrepancies between the history and the results from drug analyses concerning the identity of the drugs taken were found in 55% of the cases. In 18% of the cases, the discrepancies were considered clinically important. Serious symptoms occurred in approximately 20% of the patients, but none of them were the result of incorrect information obtained on admission. All the patients survived. These results support the prevailing view that rapid identification of the drugs taken in overdose by means of comprehensive drug screens would have little effect on the treatment of most cases of acute poisoning. However, such assays would enable optimal treatment of many cases of acute poisoning by reducing the need for supervision and costly treatments and facilitating the identification of cases that would require prompt drug-specific treatment.


Subject(s)
Drug Overdose/etiology , Medical History Taking , Acute Disease , Adult , Anti-Anxiety Agents/blood , Anti-Anxiety Agents/poisoning , Anti-Anxiety Agents/urine , Benzodiazepines , Drug Overdose/blood , Drug Overdose/diagnosis , Drug Overdose/urine , Emergency Service, Hospital , Gastric Lavage , Humans , Prospective Studies , Suicide, Attempted , Surveys and Questionnaires
17.
Clin Pharmacol Ther ; 68(5): 487-94, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11103751

ABSTRACT

OBJECTIVE: To examine the possible interaction of itraconazole with orally and intravenously administered dexamethasone. METHODS: In a randomized, double-blind, placebo-controlled crossover study with four phases, eight healthy subjects took either 200 mg itraconazole (in two phases) or placebo (in two phases) orally once daily for 4 days. On day 4 each subject received an oral dose of 4.5 mg dexamethasone or an intravenous dose of 5.0 mg dexamethasone sodium phosphate during both itraconazole and placebo phases. Plasma dexamethasone and cortisol concentrations were determined by HPLC up to 71 hours, itraconazole and hydroxyitraconazole up to 23 hours. RESULTS: Itraconazole decreased the systemic clearance of intravenously administered dexamethasone by 68% (P < .001), increased the total area under the plasma dexamethasone concentration-time curve [AUC(0-infinity)] 3.3-fold (P < .001), and prolonged the elimination half-life of dexamethasone 3.2-fold (P < .001). The AUC(0-infinity) of oral dexamethasone was increased 3.7-fold (P < .001), the peak plasma concentration 1.7-fold (P < .001), and the elimination half-life 2.8-fold (P < .001) by itraconazole. The morning plasma cortisol concentrations measured 47 and 71 hours after administration of dexamethasone were substantially lower after exposure to itraconazole than to placebo (P < .001). Accordingly, the adrenal-suppressant effect of dexamethasone was greatly enhanced during the itraconazole phases. CONCLUSIONS: Itraconazole markedly increases the systemic exposure to and effects of dexamethasone. A careful follow-up is recommended when itraconazole or other potent inhibitors of the cytochrome P450 3A4 are added to the drug regimen of patients receiving dexamethasone.


Subject(s)
Antifungal Agents/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Dexamethasone/pharmacokinetics , Glucocorticoids/pharmacokinetics , Itraconazole/pharmacology , Mixed Function Oxygenases/antagonists & inhibitors , Administration, Oral , Adult , Antifungal Agents/administration & dosage , Antifungal Agents/blood , Area Under Curve , Chromatography, High Pressure Liquid , Cross-Over Studies , Cytochrome P-450 CYP3A , Dexamethasone/administration & dosage , Dexamethasone/blood , Double-Blind Method , Drug Interactions , Female , Glucocorticoids/administration & dosage , Glucocorticoids/blood , Half-Life , Humans , Hydrocortisone/blood , Injections, Intravenous , Itraconazole/administration & dosage , Itraconazole/blood , Male , Metabolic Clearance Rate/drug effects
18.
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
19.
Clin Pharmacol Ther ; 68(4): 384-90, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11061578

ABSTRACT

BACKGROUND: Grapefruit juice is a potent inhibitor of CYP3A4-mediated drug metabolism. We wanted to investigate how long the inhibitory effect of grapefruit juice lasts, with the CYP3A4 substrate simvastatin used as a model drug. METHODS: This crossover study consisted of 5 study days, during which 10 healthy volunteers ingested 40 mg simvastatin with water (control), with "high-dose" grapefruit juice (200 mL double-strength grapefruit juice three times a day for 3 days), or 1, 3, and 7 days after ingestion of "high-dose" grapefruit juice. For safety reasons, the study was performed in three parts to allow simvastatin-free days between the study days. Serum concentrations of simvastatin and simvastatin acid were measured by liquid chromatography-tandem mass spectrometry up to 12 hours. RESULTS: When simvastatin was taken with grapefruit juice, the mean peak serum concentration (Cmax) and the mean area under the serum concentration-time curve [AUC(0-infinity)] of simvastatin were increased 12.0-fold (P < .001) and 13.5-fold (P < .001), respectively, compared with control. When simvastatin was administered 24 hours after ingestion of the last dose of grapefruit juice, the Cmax and AUC(0-infinity) were increased 2.4-fold (P < .01) and 2.1-fold (P < .001), respectively, compared with control. When simvastatin was given 3 days after ingestion of grapefruit juice, the Cmax and AUC(0-infinity) were increased 1.5-fold (P = .12) and 1.4-fold (P = .09), respectively, compared with control. Seven days after ingestion of grapefruit juice, no differences in the Cmax or AUC(0-infinity) of simvastatin were seen. The mean Cmax and AUC(0-infinity) of simvastatin acid were increased 5.0-fold and 4.5-fold, respectively (P < .001), compared with control when simvastatin was taken with grapefruit juice and 1.7-fold (P < .01) when it was taken 24 hours after ingestion of grapefruit juice. After an interval of 3 or 7 days between ingestion of grapefruit juice and simvastatin, the pharmacokinetic variables of simvastatin acid did not differ significantly from those in the control phase. CONCLUSIONS: When simvastatin is taken 24 hours after ingestion of "high-dose" grapefruit juice, the effect of grapefruit juice on the AUC of simvastatin is only about 10% of the effect observed during concomitant intake of grapefruit juice and simvastatin. The interaction potential of even high amounts of grapefruit juice with CYP3A4 substrates dissipates within 3 to 7 days after ingestion of the last dose of grapefruit juice.


Subject(s)
Citrus , Cytochrome P-450 Enzyme System/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Mixed Function Oxygenases/metabolism , Simvastatin/pharmacokinetics , Administration, Oral , Adult , Analysis of Variance , Area Under Curve , Beverages , Cross-Over Studies , Cytochrome P-450 CYP3A , Drug Administration Schedule , Female , Food-Drug Interactions , Gas Chromatography-Mass Spectrometry , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hypolipidemic Agents/pharmacokinetics , Male , Reference Values , Simvastatin/administration & dosage , Simvastatin/blood , Time Factors
20.
Eur J Clin Pharmacol ; 56(6-7): 489-93, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11049012

ABSTRACT

OBJECTIVE: To investigate whether the pharmacokinetics of orally administered methylprednisolone and plasma cortisol concentrations are affected by administration of grapefruit juice. METHODS: In a randomised, two-phase, cross-over study, ten healthy subjects received either 200 ml double-strength grapefruit juice or water three times a day for 2 days. On day 3, 16 mg methylprednisolone was given orally with 200 ml grapefruit juice or water. Additionally, 200 ml grapefruit juice or water was ingested 0.5 h and 1.5 h after methylprednisolone administration. Plasma concentrations of methylprednisolone and cortisol were determined using liquid chromatography/mass spectrometry (LC/MS/MS) over a 47-h period. RESULTS: Grapefruit juice increased the total area under the plasma methylprednisolone concentration-time curve (AUC 0--infinity) by 75% (P < 0.001) and the elimination half-life (t1/2) of methylprednisolone by 35% (P < 0.001). The peak plasma concentration of methylprednisolone (Cmax) was increased by 27% (P < 0.01). Grapefruit juice delayed the time to the Cmax from 2.0 h to 3.0 h (P < 0.05). There was no significant difference in the plasma cortisol concentrations, measured after methylprednisolone administration, between the water and grapefruit juice phases. However, grapefruit juice slightly decreased the morning plasma cortisol concentrations before methylprednisolone administration (P < 0.05). CONCLUSIONS: Grapefruit juice given in high amounts moderately increases the AUC 0--infinity and t1/2 of oral methylprednisolone. The increase in t1/2 suggests that grapefruit juice can affect the systemic methylprednisolone metabolism. The clinical significance of the grapefruit juice-methylprednisolone interaction is small, but in some sensitive subjects high doses of grapefruit juice might enhance the effects of oral methylprednisolone.


Subject(s)
Anti-Inflammatory Agents/blood , Beverages , Citrus , Food-Drug Interactions , Methylprednisolone/blood , Adult , Area Under Curve , Cross-Over Studies , Double-Blind Method , Female , Half-Life , Humans , Hydrocortisone/blood , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...