Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
1.
Pharmacogenomics J ; 18(1): 136-143, 2018 01.
Article in English | MEDLINE | ID: mdl-28290528

ABSTRACT

A common haplotype of the flavin-containing monooxygenase gene FMO3 is associated with aberrant mRNA splicing, a twofold reduction in in vivo nicotine N-oxidation and reduced nicotine dependence. Tobacco remains the largest cause of preventable mortality worldwide. CYP2A6, the primary hepatic nicotine metabolism gene, is robustly associated with cigarette consumption but other enzymes contribute to nicotine metabolism. We determined the effects of common variants in FMO3 on plasma levels of nicotine-N-oxide in 170 European Americans administered deuterated nicotine. The polymorphism rs2266780 (E308G) was associated with N-oxidation of both orally administered and ad libitum smoked nicotine (P⩽3.3 × 10-5 controlling for CYP2A6 genotype). In vitro, the FMO3 G308 variant was not associated with reduced activity, but rs2266780 was strongly associated with aberrant FMO3 mRNA splicing in both liver and brain (P⩽6.5 × 10-9). Surprisingly, in treatment-seeking European American smokers (n=1558) this allele was associated with reduced nicotine dependence, specifically with a longer time to first cigarette (P=9.0 × 10-4), but not with reduced cigarette consumption. As N-oxidation accounts for only a small percentage of hepatic nicotine metabolism we hypothesized that FMO3 genotype affects nicotine metabolism in the brain (unlike CYP2A6, FMO3 is expressed in human brain) or that nicotine-N-oxide itself has pharmacological activity. We demonstrate for the first time nicotine N-oxidation in human brain, mediated by FMO3 and FMO1, and show that nicotine-N-oxide modulates human α4ß2 nicotinic receptor activity in vitro. These results indicate possible mechanisms for associations between FMO3 genotype and smoking behaviors, and suggest nicotine N-oxidation as a novel target to enhance smoking cessation.


Subject(s)
Brain/metabolism , Nicotine/adverse effects , Nicotine/metabolism , Oxygenases/genetics , Oxygenases/metabolism , Polymorphism, Genetic/genetics , Tobacco Use Disorder/genetics , Alleles , Animals , Cells, Cultured , Genotype , Haplotypes/genetics , Humans , Male , Middle Aged , Oocytes/metabolism , Oxidation-Reduction , Smoking/genetics , Smoking/metabolism , Tobacco Use Disorder/metabolism , White People , Xenopus/genetics
4.
Clin Pharmacol Ther ; 95(4): 376-82, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24458010

ABSTRACT

Codeine is bioactivated to morphine, a strong opioid agonist, by the hepatic cytochrome P450 2D6 (CYP2D6); hence, the efficacy and safety of codeine are governed by CYP2D6 activity. Polymorphisms are a major cause of CYP2D6 variability. We summarize evidence from the literature supporting this association and provide therapeutic recommendations for codeine based on CYP2D6 genotype. This document is an update to the 2012 Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for CYP2D6 genotype and codeine therapy.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Codeine/pharmacokinetics , Cytochrome P-450 CYP2D6/genetics , Pharmacogenetics , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Codeine/administration & dosage , Codeine/adverse effects , Cytochrome P-450 CYP2D6/metabolism , Genetic Testing , Genotype , Humans , Morphine/metabolism , Polymorphism, Genetic
5.
Clin Pharmacol Ther ; 94(3): 394-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23665868

ABSTRACT

Metoprolol, a commonly prescribed ß-blocker, is primarily metabolized by cytochrome P450 2D6 (CYP2D6), an enzyme with substantial genetic heterogeneity. Several smaller studies have shown that metoprolol pharmacokinetics is influenced by CYP2D6 genotype and metabolizer phenotype. To increase robustness of metoprolol pharmacokinetic estimates, a systematic review and meta-analysis of pharmacokinetic studies that administered a single oral dose of immediate-release metoprolol were performed. Pooled analysis (n = 264) demonstrated differences in peak plasma metoprolol concentration, area under the concentration-time curve, elimination half-life, and apparent oral clearance that were 2.3-, 4.9-, 2.3-, and 5.9-fold between extensive and poor metabolizers, respectively, and 5.3-, 13-, 2.6-, and 15-fold between ultrarapid and poor metabolizers (all P < 0.001), respectively. Enantiomer-specific analysis revealed genotype-dependent enantio-selective metabolism, with nearly 40% greater R- than S-metoprolol metabolism in ultrarapid and extensive metabolizers. This study demonstrates a marked effect of CYP2D6 metabolizer phenotype on metoprolol pharmacokinetics and confirms enantiomer-specific metabolism of metoprolol.


Subject(s)
Adrenergic beta-Antagonists/pharmacokinetics , Cytochrome P-450 CYP2D6/metabolism , Metoprolol/pharmacokinetics , Adrenergic beta-Antagonists/chemistry , Cytochrome P-450 CYP2D6/genetics , Gene Dosage , Humans , Metoprolol/chemistry , Phenotype , Stereoisomerism
6.
Clin Pharmacol Ther ; 93(5): 402-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23486447

ABSTRACT

Polymorphisms in CYP2D6 and CYP2C19 affect the efficacy and safety of tricyclics, with some drugs being affected by CYP2D6 only, and others by both polymorphic enzymes. Amitriptyline, clomipramine, doxepin, imipramine, and trimipramine are demethylated by CYP2C19 to pharmacologically active metabolites. These drugs and their metabolites, along with desipramine and nortriptyline, undergo hydroxylation by CYP2D6 to less active metabolites. Evidence from published literature is presented for CYP2D6 and CYP2C19 genotype-directed dosing of tricyclic antidepressants.


Subject(s)
Antidepressive Agents, Tricyclic/administration & dosage , Aryl Hydrocarbon Hydroxylases/genetics , Cytochrome P-450 CYP2D6/genetics , Antidepressive Agents, Tricyclic/adverse effects , Antidepressive Agents, Tricyclic/pharmacokinetics , Cytochrome P-450 CYP2C19 , Dose-Response Relationship, Drug , Genotype , Humans , Mental Disorders/drug therapy , Mental Disorders/physiopathology , Pharmacogenetics , Polymorphism, Genetic
7.
Clin Pharmacol Ther ; 91(4): 673-84, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22398970

ABSTRACT

Mechanisms by which efavirenz diminishes methadone plasma concentrations are unknown. This investigation determined efavirenz influence on clinical methadone disposition and miosis, intravenous and oral alfentanil clearance (hepatic and intestinal cytochrome P450 3A4/5 (CYP3A4/5) activity), fexofenadine disposition (intestinal transporters activity), and efavirenz clearance and 8-hydroxylation (CYP2B6 activity), and human hepatocyte effects. Efavirenz induced systemic and oral alfentanil clearances two- to fivefold and induced efavirenz 8-hydroxylation. Efavirenz stereoselectively decreased methadone plasma concentrations 50-70%. Methadone systemic and oral clearances, hepatic clearance and extraction ratio, N-demethylation, and metabolite formation clearance were stereoselectively increased two- to threefold. Bioavailability decreased. Efavirenz shifted methadone concentration-miosis curves leftward and upward. Efavirenz induced hepatocyte CYP2B6 and CYP3A4 expression, activity, and methadone N-demethylation. Results show that efavirenz coinduced hepatic CYP2B6 and CYP3A4/5, coinduced hepatic and intestinal CYP3A4/5, and coinduced gastrointestinal CYP3A4/5 and efflux transporters. Methadone disposition was most consistent with efavirenz induction of hepatic CYP2B6-mediated methadone N-demethylation. Efavirenz may alter methadone pharmacodynamics.


Subject(s)
Benzoxazines/blood , Benzoxazines/pharmacokinetics , Methadone/blood , Methadone/pharmacokinetics , Adolescent , Adult , Alkynes , Aryl Hydrocarbon Hydroxylases/metabolism , Benzoxazines/pharmacology , Cross-Over Studies , Cyclopropanes , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP3A/metabolism , Drug Interactions/physiology , Female , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Male , Methadone/pharmacology , Oxidoreductases, N-Demethylating/metabolism , Young Adult
8.
Clin Pharmacol Ther ; 91(2): 321-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22205192

ABSTRACT

Codeine is bioactivated to morphine, a strong opioid agonist, by the hepatic cytochrome P450 2D6 (CYP2D6); hence, the efficacy and safety of codeine as an analgesic are governed by CYP2D6 polymorphisms. Codeine has little therapeutic effect in patients who are CYP2D6 poor metabolizers, whereas the risk of morphine toxicity is higher in ultrarapid metabolizers. The purpose of this guideline (periodically updated at http://www.pharmgkb.org) is to provide information relating to the interpretation of CYP2D6 genotype test results to guide the dosing of codeine.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Codeine/administration & dosage , Codeine/pharmacokinetics , Cytochrome P-450 CYP2D6/genetics , Pharmacogenetics/standards , Analgesics, Opioid/adverse effects , Drug Dosage Calculations , Genetic Testing/standards , Humans , Phenotype , Polymorphism, Genetic
9.
Clin Pharmacol Ther ; 90(1): 100-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21562488

ABSTRACT

Systemic and oral clearances of alfentanil (ALF) are in vivo probes for hepatic and first-pass cytochrome P450 (CYP) 3A. Both ALF single-point plasma concentrations and miosis are surrogates for area under the concentration-time curve (AUC) and clearance and are minimal and noninvasive CYP3A probes. This investigation determined ALF sensitivity for detecting graded CYP3A induction and compared it with that of midazolam (MDZ). Twelve volunteers (sequential crossover) received 0, 5, 10, 25, or 75 mg oral rifampin for 5 days. MDZ and ALF were given intravenously and orally on sequential days. Dark-adapted pupil diameter was measured with blood sampling. Graded rifampin decreased plasma MDZ AUCs to 83, 76, 62, and 59% (intravenous (i.v.)) and 78, 66, 39, and 24% (oral) of control. Hepatic and first-pass CYP3A induction were detected comparably by plasma MDZ and ALF AUCs. Single ALF concentrations detected all CYP3A induction, whereas MDZ was less sensitive. ALF miosis detected induction of first-pass but not hepatic CYP3A.


Subject(s)
Alfentanil/pharmacology , Analgesics, Opioid/pharmacology , Cytochrome P-450 CYP3A/biosynthesis , Liver/drug effects , Liver/enzymology , Miosis/chemically induced , Administration, Oral , Adolescent , Adult , Alfentanil/administration & dosage , Analgesics, Opioid/administration & dosage , Area Under Curve , Biological Availability , Cross-Over Studies , Enzyme Induction/drug effects , Female , Half-Life , Humans , Hypnotics and Sedatives/blood , Hypnotics and Sedatives/pharmacology , Injections, Intravenous , Kinetics , Leprostatic Agents/pharmacology , Male , Midazolam/blood , Midazolam/pharmacology , Rifampin/pharmacology , Smoking/metabolism , Young Adult
10.
Clin Pharmacol Ther ; 89(4): 562-70, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21346758

ABSTRACT

Alfentanil (ALF) is a validated probe for hepatic, first-pass, and intestinal cytochrome P450 (CYP) 3A activity, using plasma clearances, single-point concentrations, and noninvasive pupil diameter change (miosis). Assessing intravenous (i.v.) and oral drug disposition typically requires separate dosing. This investigation evaluated concurrent administration of oral deuterated and i.v. unlabeled ALF to assess both intestinal and hepatic CYP3A, and compare sequential and simultaneous dosing. ALF disposition was evaluated after strong hepatic and/or intestinal CYP3A induction and inhibition by rifampin, ketoconazole, and grapefruit juice. Using plasma ALF concentrations and area under the curve (AUC), clearance, or single-point concentrations, both simultaneous and sequential dosing provided equivalent results and detected hepatic and intestinal CYP3A induction and inhibition. Miosis better detected CYP3A modulation with sequential vs. simultaneous dosing. These results show that concurrent administration of oral deuterated and i.v. ALF, either sequentially or simultaneously, is an efficient and effective approach to assessing hepatic and intestinal CYP3A activity.


Subject(s)
Alfentanil/pharmacokinetics , Anesthetics, Intravenous/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Miosis/chemically induced , Administration, Oral , Adult , Alfentanil/administration & dosage , Alfentanil/pharmacology , Anesthetics, Intravenous/administration & dosage , Anesthetics, Intravenous/pharmacology , Area Under Curve , Beverages , Citrus paradisi/chemistry , Cross-Over Studies , Cytochrome P-450 CYP3A/drug effects , Deuterium , Drug Administration Schedule , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Female , Humans , Intestinal Mucosa/metabolism , Ketoconazole/pharmacology , Liver/metabolism , Male , Rifampin/pharmacology , Young Adult
11.
Clin Pharmacol Ther ; 84(1): 158-62, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18509326

ABSTRACT

Problems with organ toxicity from the halogenated volatile anesthetics have influenced clinical practice, anesthetic selection, and drug development for half a century.(1,2) In turn, investigations that elucidated the mechanisms of volatile anesthetics adverse effects identified new mechanisms, provided remarkable insights into human toxicology, and represented seminal contributions to clinical pharmacology. This review focuses on adverse organ effects (hepatic, renal, and others) that are attributable to anesthetic metabolism and/or degradation. Routine reversible effects on pulmonary, cardiac, and other organ functions are not addressed.


Subject(s)
Anesthetics, Inhalation/adverse effects , Halothane/adverse effects , Anesthetics, Inhalation/pharmacokinetics , Halothane/pharmacokinetics , Humans , Hydrocarbons, Halogenated/adverse effects , Hydrocarbons, Halogenated/pharmacokinetics , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism
12.
Clin Pharmacol Ther ; 84(4): 497-505, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19238655

ABSTRACT

Ritonavir diminishes methadone plasma concentrations, an effect attributed to CYP3A induction, but the actual mechanisms are unknown. We determined ritonavir effects on stereoselective methadone pharmacokinetics and clinical effects (pupillary miosis) in healthy human immunodeficiency virus-negative volunteers. Subjects received intravenous plus oral (deuterium-labeled) racemic methadone after no ritonavir, short-term (3-day) ritonavir, and steady-state ritonavir. Acute and steady-state ritonavir, respectively, caused 1.5- and 2-fold induction of systemic and apparent oral R- and S-methadone clearances. Ritonavir increased renal clearance 40-50%, and stereoselectively (S > R) increased hepatic methadone N-demethylation 50-80%, extraction twofold, and clearance twofold. Bioavailability was unchanged despite significant inhibition of intestinal P-glycoprotein. Intestinal and hepatic CYP3A was inhibited > 70%. Ritonavir shifted methadone plasma concentration-miosis curves leftward and upward. Rapid ritonavir induction of methadone clearance results from increased renal clearance and induced hepatic metabolism. Induction of methadone metabolism occurred despite profound CYP3A inhibition, suggesting no role for CYP3A in clinical methadone metabolism and clearance. Ritonavir may alter methadone pharmacodynamics.


Subject(s)
Cytochrome P-450 CYP3A/physiology , HIV Protease Inhibitors/pharmacology , Methadone/pharmacokinetics , Narcotics/pharmacokinetics , Ritonavir/pharmacology , Adult , Area Under Curve , Biological Availability , Cross-Over Studies , Cytochrome P-450 CYP3A Inhibitors , Dose-Response Relationship, Drug , Drug Interactions , Female , HIV Protease Inhibitors/pharmacokinetics , Humans , Male , Methadone/pharmacology , Narcotics/pharmacology , Pupil/drug effects , Ritonavir/pharmacokinetics , Stereoisomerism
13.
Clin Pharmacol Ther ; 84(4): 506-12, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19238656

ABSTRACT

Ritonavir diminishes methadone plasma concentrations, an effect attributed to CYP3A induction, but the actual mechanisms are unknown. We determined short-term (2-day) and steady-state (2-week) ritonavir effects on intestinal and hepatic CYP3A4/5 (probed with intravenous (IV) and oral alfentanil (ALF) and with miosis) and P-glycoprotein (P-gp) (fexofenadine), and on methadone pharmacokinetics and pharmacodynamics in healthy volunteers. Acute ritonavir increased the area under the concentration-time curve (AUC)(0-infinity)/dose ratio (ritonavir/control) for oral ALF 25-fold. Steady-state ritonavir increased the AUC(0-Infinity)/dose ratio for IV and oral ALF 4- and 10-fold, respectively; reduced hepatic extraction (from 0.26 to 0.07) and intestinal extraction (from 0.51 to 0); and increased bioavailability (from 37 to 95%). Acute ritonavir inhibits first-pass CYP3A > 96%. Chronic ritonavir inhibits hepatic CYP3A (> 70%) and first-pass CYP3A (> 90%). Acute and steady-state ritonavir increased the fexofenadine AUC(0-infinity) 2.8- and 1.4-fold, respectively, suggesting P-gp inhibition. Steady-state compared with acute ritonavir caused mild apparent induction of P-gp and hepatic CYP3A, but net inhibition still predominated. Ritonavir inhibited both intestinal and hepatic CYP3A and drug transport. ALF miosis noninvasively determined CYP3A inhibition by ritonavir.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cytochrome P-450 CYP3A/physiology , HIV Protease Inhibitors/pharmacology , Methadone/pharmacokinetics , Narcotics/pharmacokinetics , Ritonavir/pharmacology , Adult , Alfentanil/administration & dosage , Alfentanil/blood , Alfentanil/pharmacokinetics , Area Under Curve , Biological Availability , Cross-Over Studies , Cytochrome P-450 CYP3A Inhibitors , Dose-Response Relationship, Drug , Drug Interactions , Female , HIV Protease Inhibitors/pharmacokinetics , Humans , Intestines/enzymology , Liver/enzymology , Male , Methadone/pharmacology , Narcotics/pharmacology , Pupil/drug effects , Ritonavir/pharmacokinetics , Stereoisomerism , Terfenadine/administration & dosage , Terfenadine/analogs & derivatives , Terfenadine/blood , Terfenadine/pharmacokinetics
14.
Clin Pharmacol Ther ; 83(1): 77-85, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17495874

ABSTRACT

Itraconazole (ITZ) is metabolized in vitro to three inhibitory metabolites: hydroxy-itraconazole (OH-ITZ), keto-itraconazole (keto-ITZ), and N-desalkyl-itraconazole (ND-ITZ). The goal of this study was to determine the contribution of these metabolites to drug-drug interactions caused by ITZ. Six healthy volunteers received 100 mg ITZ orally for 7 days, and pharmacokinetic analysis was conducted at days 1 and 7 of the study. The extent of CYP3A4 inhibition by ITZ and its metabolites was predicted using this data. ITZ, OH-ITZ, keto-ITZ, and ND-ITZ were detected in plasma samples of all volunteers. A 3.9-fold decrease in the hepatic intrinsic clearance of a CYP3A4 substrate was predicted using the average unbound steady-state concentrations (C(ss,ave,u)) and liver microsomal inhibition constants for ITZ, OH-ITZ, keto-ITZ, and ND-ITZ. Accounting for circulating metabolites of ITZ significantly improved the in vitro to in vivo extrapolation of CYP3A4 inhibition compared to a consideration of ITZ exposure alone.


Subject(s)
Antifungal Agents/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Itraconazole/analogs & derivatives , Itraconazole/pharmacology , Liver/drug effects , Administration, Oral , Adult , Antifungal Agents/administration & dosage , Antifungal Agents/pharmacokinetics , Biotransformation , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Female , Glucuronides/metabolism , Humans , Itraconazole/administration & dosage , Itraconazole/pharmacokinetics , Liver/enzymology , Male , Models, Biological
15.
Clin Pharmacol Ther ; 82(4): 410-26, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17554244

ABSTRACT

The hepatic and first-pass cytochrome P4503A (CYP3A) probe alfentanil (ALF) is also metabolized in vitro by CYP3A5. Human hepatic microsomal ALF metabolism is higher in livers with at least one CYP3A5*1 allele and higher CYP3A5 protein content, compared with CYP3A5*3 homozygotes with little CYP3A5. The influence of CYP3A5 genotype on ALF pharmacokinetics and pharmacodynamics was studied, and compared to midazolam (MDZ), another CYP3A probe. Healthy volunteers (58 men, 41 women) were genotyped for CYP3A5 *1, *3, *6, and *7 alleles. They received intravenous MDZ then ALF, and oral MDZ and ALF the next day. Plasma MDZ and ALF concentrations were determined by mass spectrometry. Dark-adapted pupil diameters were determined coincident with blood sampling. In CYP3A5(*)3/(*)3 (n=62), (*)1/(*)3 (n=28), and (*)1/(*)1 (n=8) genotypes, systemic clearances of ALF were 4.6+/-1.8, 4.8+/-1.7, and 3.9+/-1.7 ml/kg/min and those of MDZ were 7.8+/-2.3, 7.7+/-2.3, and 6.0+/-1.4 ml/kg/min, respectively (not significant), and apparent oral clearances were 11.8+/-7.2, 13.3+/-6.1, and 12.6+/-8.2 ml/kg/min for ALF and 35.2+/-19.0, 36.4+/-15.7, and 29.4+/-9.3 ml/kg/min for MDZ (not significant). Clearances were not different between African Americans (n=25) and Whites (n=68), or between CYP3A5 genotypes within African Americans. ALF pharmacodynamics was not different between CYP3A5 genotypes. There was consistent concordance between ALF and MDZ, in clearances and extraction ratios. Thus, in a relatively large cohort of healthy subjects with constitutive CYP3A activity, CYP3A5 genotype had no effect on the systemic or apparent oral clearances, or pharmacodynamics, of the CYP3A probes ALF and MDZ, despite affecting their hepatic microsomal metabolism.


Subject(s)
Alfentanil/pharmacokinetics , Cytochrome P-450 Enzyme System/genetics , Midazolam/pharmacokinetics , Polymorphism, Genetic , Administration, Oral , Adult , Black or African American/genetics , Alfentanil/administration & dosage , Alfentanil/adverse effects , Alfentanil/blood , Biomarkers/metabolism , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Female , Gene Frequency , Genotype , Hispanic or Latino/genetics , Humans , Injections, Intravenous , Male , Midazolam/administration & dosage , Midazolam/blood , Middle Aged , Miosis/chemically induced , Phenotype , Pupil/drug effects , Reference Values , Substrate Specificity , White People/genetics
16.
Acta Anaesthesiol Scand ; 47(6): 765-70, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12803597

ABSTRACT

BACKGROUND: Cytochrome P4502E1(CYP2E1)-mediated oxidation of halothane to a reactive intermediate (trifluoroacyl chloride) that covalently binds to hepatic proteins forming trifluoroacetylated neoantigens is believed to be the initiating event in a complex immunologic cascade culminating in antibody formation and severe hepatic necrosis ('halothane hepatitis') in susceptible patients. Trifluoroacyl chloride may also hydrolyze to the stable metabolite trifluoroacetic acid (TFA). CYP2E1 inactivation by disulfiram or its primary metabolite, diethyldithiocarbamate, inhibits human halothane oxidation to TFA in vitro and in vivo. Nevertheless, disulfiram effects on hepatic protein trifluoroacetylation by halothane in vivo are unknown. This investigation tested the hypotheses that disulfiram prevents halothane-dependent protein trifluoroacetylation in vivo, and that TFA represents a biomarker for hepatic protein trifluoroacetylation. METHODS: Rats were pretreated with isoniazid (CYP2E1 induction), isoniazid followed by disulfiram (CYP2E1 inhibition), or nothing (controls), then anesthetized with halothane or nothing (controls). Plasma and urine TFA were quantified by ion HPLC; hepatic microsomal TFA-proteins were analyzed by Western blot. RESULTS: CYP2E1 induction increased both TFA and TFA-protein formation compared with uninduced halothane-treated rats. Disulfiram, even after CYP2E1 induction, nearly abolished both TFA and TFA-protein formation. Pretreatments similarly affected both TFA and TFA-protein formation across all groups. CONCLUSIONS: Disulfiram inhibition of CYP2E1-mediated halothane oxidation prevents hepatic protein trifluoroacetylation. Based on the concordance between TFA and TFA-protein formation, TFA appears to be a valid biomarker for TFA-protein formation. Disulfiram inhibition of human halothane oxidation in vivo, previously assessed by diminished TFA formation, probably also confers inhibition of hepatic TFA-protein formation.


Subject(s)
Anesthetics, Inhalation/metabolism , Disulfiram/pharmacology , Halothane/metabolism , Liver/metabolism , Proteins/metabolism , Trifluoroacetic Acid/metabolism , Animals , Blotting, Western , Bromides/metabolism , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2E1 Inhibitors , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Isoniazid/pharmacology , Liver/drug effects , Male , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
17.
Acta Anaesthesiol Scand ; 47(3): 241-59, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12648189

ABSTRACT

BACKGROUND: The gender aspect in pharmacokinetics and pharmacodynamics of anesthetics has attracted little attention. Knowledge of previous work is required to decide if gender-based differences in clinical practice is justified, and to determine the need for research. METHODS: Basis for this paper was obtained by Medline searches using the key words 'human' and 'gender' or 'sex,' combined with individual drug names. The reference lists of these papers were further checked for other relevant studies. RESULTS: Females have 20-30% greater sensitivity to the muscle relaxant effects of vecuronium, pancuronium and rocuronium. When rapid onset of or short duration of action is very important, gender-modified dosing may be considered. Males are more sensitive than females to propofol. It may therefore be necessary to decrease the propofol dose by 30-40% in males compared with females in order to achieve similar recovery times. Females are more sensitive than males to opioid receptor agonists, as shown for morphine as well as for a number of kappa (OP2) receptor agonists. On this basis, males will be expected to require 30-40% higher doses of opioid analgesics than females to achieve similar pain relief. On the other hand, females may experience respiratory depression and other adverse effects more easily if they are given the same doses as males. CONCLUSION: These examples illustrate that gender should be taken into account as a factor that may be predictive for the dosage of several anesthetic drugs. Moreover, there is an obvious need for more research in this area in order to further optimize drug treatment in anesthesia.


Subject(s)
Anesthetics/pharmacology , Sex Characteristics , Adrenergic Agents/pharmacokinetics , Adrenergic Agents/pharmacology , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/pharmacology , Anesthetics/pharmacokinetics , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Female , Humans , Male , Steroids
18.
Acta Anaesthesiol Scand ; 46(7): 759-70, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12139528

ABSTRACT

BACKGROUND: Nasal administration of opioids may be an alternative route to intravenous, subcutaneous, oral transmucosal, oral or rectal administration in some patients. Key features may be self-administration, combined with rapid onset of action. The aim of this paper is to evaluate the present base of knowledge on this topic. METHODS: The review is based on human studies found in Medline or in the reference list of these papers. The physiology of the nasal mucosa and some pharmaceutical aspects of nasal administration are described. The design of each study is described, but not systematically evaluated. RESULTS: Pharmacokinetic studies in volunteers are reported for fentanyl, alfentanil, sufentanil, butorphanol, oxycodone and buprenorphine. Mean times for achieving maximum serum concentrations vary from 5 to 50 min, while mean figures for bioavailability vary from 46 to 71%. Fentanyl, pethidine and butorphanol have been studied for postoperative pain. Mean onset times vary from 12 to 22 min and times to peak effect from 24 to 60 min. There is considerable interindividual variation in pharmacokinetics and clinical outcome. This may partly be due to lack of optimization of nasal formulations. Patient-controlled nasal analgesia is an effective alternative to intravenous PCA. Adverse effects are mainly those related to the opioids themselves, rather than to nasal administration. Some experience with nasal opioids in outpatients and for chronic pain has also been reported. CONCLUSION: Nasal administration of opioids has promising features, but is still in its infancy. Adequately designed clinical studies are needed. Improvements of nasal sprayer devices and opioid formulations may improve clinical outcome.


Subject(s)
Analgesics, Opioid/administration & dosage , Pain/drug therapy , Administration, Intranasal , Ambulatory Surgical Procedures , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacokinetics , Chronic Disease , Humans , Nasal Mucosa/metabolism , Pain, Postoperative/drug therapy
19.
Anesth Analg ; 93(6): 1511-20, table of contents, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11726433

ABSTRACT

UNLABELLED: Sevoflurane degradation by carbon dioxide absorbents during low-flow anesthesia forms the haloalkene Compound A, which causes nephrotoxicity in rats. Numerous studies have shown no effects of Compound A formation on postoperative renal function after moderate-duration (3-4 h) low-flow sevoflurane; however, effects of longer exposures remain unresolved. We compared renal function after long-duration low-flow (<1 L/min) sevoflurane and isoflurane anesthesia in consenting surgical patients with normal renal function. To maximize degradant exposure, Baralyme was used, and anesthetic concentrations were maximized (no nitrous oxide and minimal opioids). Inspired and expired Compound A concentrations were quantified. Blood and urine were obtained for laboratory evaluation. Sevoflurane (n = 28) and isoflurane (n = 27) groups were similar with respect to age, sex, weight, ASA status, and anesthetic duration (9.1 +/- 3.0 and 8.2 +/- 3.0 h, mean +/- SD) and exposure (9.2 +/- 3.6 and 9.1 +/- 3.7 minimum alveolar anesthetic concentration hours). Maximum inspired Compound A was 25 +/- 9 ppm (range, 6-49 ppm), and exposure (area under the concentration-time curve) was 165 +/- 95 (35-428) ppm. h. There was no significant difference between anesthetic groups in 24- or 72-h serum creatinine, blood urea nitrogen, creatinine clearance, or 0- to 24-h or 48- to 72-h urinary protein or glucose excretion. Proteinuria and glucosuria were common in both groups. There was no correlation between Compound A exposure and any renal function measure. There was no difference between anesthetic groups in 24- or 72-h aspartate aminotransferase or alanine aminotransferase. These results show that the renal and hepatic effects of long-duration low-flow sevoflurane and isoflurane were similar. No evidence for low-flow sevoflurane nephrotoxicity was observed, even at high Compound A exposures as long as 17 h. Proteinuria and glucosuria were common and nonspecific postoperative findings. Long-duration low-flow sevoflurane seems as safe as long-duration low-flow isoflurane anesthesia. IMPLICATIONS: Postoperative renal function after long-duration low-flow sevoflurane (with Compound A exposures greater than those typically reported) and isoflurane anesthesia were not different, as assessed by serum creatinine, blood urea nitrogen, and urinary excretion of protein and glucose. This suggests that low-flow sevoflurane is as safe as low-flow isoflurane, even at long exposures.


Subject(s)
Anesthetics, Inhalation/administration & dosage , Isoflurane/administration & dosage , Kidney/drug effects , Liver/drug effects , Methyl Ethers/administration & dosage , Adult , Aged , Aged, 80 and over , Alanine Transaminase/blood , Anesthetics, Inhalation/analysis , Aspartate Aminotransferases/blood , Breath Tests , Ethers/analysis , Female , Fluorides/blood , Humans , Hydrocarbons, Fluorinated/analysis , Kidney/physiology , Liver/physiology , Male , Middle Aged , Sevoflurane , Time Factors
20.
Clin Pharmacol Ther ; 70(6): 505-17, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11753266

ABSTRACT

OBJECTIVE: The opioid alfentanil is a CYP3A4 substrate whose plasma clearance depends exclusively on hepatic CYP3A4 activity. Alfentanil clearance is an excellent in vivo probe for hepatic CYP3A4 activity and drug interactions in humans. However, such pharmacokinetic studies are invasive and time-consuming, and they require extensive analytical effort. This investigation tested the hypothesis that alfentanil-induced miosis (drug effect) can be used as a surrogate measure for alfentanil plasma concentrations and that alfentanil effect clearance will reflect plasma clearance; thus alfentanil can serve as a noninvasive probe for hepatic CYP3A4. METHODS: Six healthy volunteers were studied in a 3-way randomized crossover design. Each volunteer received 1 mg intravenous midazolam, followed 1 hour later by 15 microg/kg intravenous alfentanil, after CYP3A4 induction (rifampin [INN, rifampicin]), CYP3A4 inhibition (troleandomycin), and control. Dark-adapted pupil diameter and dynamic light response were measured coincident with venous blood sampling for up to 8 hours. Midazolam and alfentanil were quantified by gas chromatography-mass spectrometry. Plasma concentrations of alfentanil and midazolam (an additional CYP3A4 probe) and pupil diameter versus time data were analyzed by use of noncompartmental modeling. Pupil diameter change was analyzed analogously to determine the area under the alfentanil effect (miosis)-time curve (AUEC), effect clearance (CL(miosis)), and effect half-time. RESULTS: Compared with control, CYP3A4 induction and inhibition significantly altered the clearances of alfentanil (2.8 +/- 1.4, 5.3 +/- 1.0, and 0.42 +/- 0.1 ml/kg/min, respectively; P <.05 versus control) and midazolam. Dark-adapted resting diameter (in millimeters) was the best measure of alfentanil pupil effects. Alfentanil-dependent miosis was significantly altered by CYP3A4 modulation, and log(diameter(0) - diameter(t)) versus time curves resembled alfentanil plasma disposition. AUEC(infinity) values after control, CYP3A4 induction, and inhibition were 280 +/- 150, 120 +/- 22, and 1030 +/- 240 mm x min, respectively (P <.05 versus control). Effect clearances (CL(miosis)) were 4.2 +/- 1.3, 8.8 +/- 2.4, and 1.2 +/- 0.8 microg/mm x min, respectively, and effect half-times were 62 +/- 23, 34 +/- 27, and 211 +/- 35 minutes, respectively (P <.05 versus control). CL(miosis) was significantly correlated with plasma clearances of alfentanil (r = 0.77, P <.001) and midazolam (r = 0.80; P <.001). CONCLUSIONS: Alfentanil effect (miosis) may be a sensitive and reliable surrogate for plasma alfentanil concentrations. Alfentanil effect kinetics may be used as a noninvasive surrogate for conventional pharmacokinetics. CL(miosis) appears to be a suitable noninvasive in vivo probe for hepatic CYP3A4 activity, and it merits further investigation.


Subject(s)
Alfentanil , Analgesics, Opioid , Liver/enzymology , Miosis/chemically induced , Adult , Alfentanil/pharmacokinetics , Analgesics, Opioid/pharmacokinetics , Antibiotics, Antitubercular/pharmacokinetics , Area Under Curve , Cross-Over Studies , Female , GABA Modulators/pharmacokinetics , Half-Life , Humans , Liver/drug effects , Male , Midazolam/pharmacokinetics , Pilot Projects , Pupil/drug effects , Rifampin/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...