Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
J Chromatogr B Analyt Technol Biomed Life Sci ; 877(27): 3003-11, 2009 Oct 01.
Article En | MEDLINE | ID: mdl-19660994

A method for identifying the metabolites of sibutramine 1-(4(chlorophenyl)-N,N-dimethyl-alpha-(2-methylpropyl))cyclobutanemethanamine) in urine, utilizing a double derivatization strategy, with N-methyl-N-(trimethylsilyl)-trifluoroacetamide and N-methyl-bis-(trifluoroacetamide), in gas chromatography/mass spectrometry is proposed. This methodology results in mass spectra with at least three fragments in abundance superior to 20%, attending the World Anti-Doping Agency identification criteria for qualitative assays. The characterization of the derivatives was obtained through two ionization modes: Chemical Ionization and Electron Impact ionization, both in full scan mode. Sibutramine was administered to 5 (five) volunteers and the excretion profile followed for 92h. Routine analytical, hydroxy-cyclobutane-bis-nor-sibutramine which becomes the more abundant metabolite in the first 10h and hydroxy-isopropyl-bis-nor-sibutramine which becomes the most abundant after 40h, were proposed for doping monitoring.


Cyclobutanes/urine , Fluoroacetates , Trimethylsilyl Compounds/urine , Acetamides , Cyclobutanes/chemistry , Cyclobutanes/metabolism , Doping in Sports , Drug Stability , Female , Gas Chromatography-Mass Spectrometry/methods , Humans , Male , Reproducibility of Results , Sensitivity and Specificity , Trifluoroacetic Acid/urine
2.
Forensic Sci Int ; 129(2): 104-9, 2002 Sep 26.
Article En | MEDLINE | ID: mdl-12243878

We describe a case of massive hemorrhage in the cerebral ventricles, probably caused by methamphetamine abuse. A 44-year-old male was found dead in a prone position in a hotel room. Old and new injection marks were observed in his right cubital fossa. Petechiae were observed on the conjunctiva of his right eye, laryngeal mucosa, epicardium and under the capsule of the liver (to a slight or moderate degree). The brain, weighing 1.67 kg, was heavily edematous; the lateral and fourth ventricles were filled with hematomas. Subarachnoid, intracerebral hemorrhages were not observed. Cerebral vascular abnormalities were not evident. There were no remarkable changes in other organs, other than congestion. Gas chromatographic-mass spectrometric analysis of the urine disclosed the presence of methamphetamine and amphetamine. The concentration of methamphetamine within the femoral venous blood and intraventricular hematoma was 0.347 microg/ml and 0.189 microg/g, respectively. Amphetamine was not detected in either sample. Urine contained 3.15 microg/ml methamphetamine and 0.063 microg/ml amphetamine. These results indicate that intraventricular hemorrhage might have occurred shortly after intravenous self-administration of methamphetamine. Cerebral arterial spasm and hypertension resulting from the administration of methamphetamine might have resulted in intraventricular hemorrhage.


Central Nervous System Stimulants/adverse effects , Cerebral Hemorrhage , Cerebral Ventricles/drug effects , Fluoroacetates , Methamphetamine/adverse effects , Acetic Anhydrides , Adult , Amphetamine/adverse effects , Amphetamine/blood , Amphetamine/urine , Central Nervous System Stimulants/blood , Central Nervous System Stimulants/urine , Fatal Outcome , Gas Chromatography-Mass Spectrometry , Humans , Male , Methamphetamine/blood , Methamphetamine/urine , Substance Abuse, Intravenous , Trifluoroacetic Acid/urine
3.
Eur J Clin Pharmacol ; 55(11-12): 853-9, 2000.
Article En | MEDLINE | ID: mdl-10805064

OBJECTIVE: Halothane undergoes both oxidative and reductive metabolism by cytochrome P450 (CYP), respectively causing rare immune-mediated hepatic necrosis and common, mild subclinical hepatic toxicity. Halothane also causes lipid peroxidation in rodents in vitro and in vivo, but in vivo effects in humans are unknown. In vitro investigations have identified a role for human CYPs 2E1 and 2A6 in oxidation and CYPs 2A6 and 3A4 in reduction. The mechanism-based CYP2E1 inhibitor disulfiram diminished human halothane oxidation in vivo. This investigation tested the hypotheses that halothane causes lipid peroxidation in humans in vivo, and that CYP2A6 or CYP3A4 inhibition can diminish halothane metabolism. METHODS: Patients (n = 9 each group) received single doses of the mechanism-based inhibitors troleandomycin (CYP3A4), methoxsalen (CYP2A6) or nothing (controls) before a standard halothane anaesthetic. Reductive halothane metabolites chlorotrifluoroethane and chlorodifluoroethylene in exhaled breath, fluoride in urine, and oxidative metabolites trifluoroacetic acid and bromide in urine were measured for 48 h postoperatively. Lipid peroxidation was assessed by plasma F2-isoprostane concentrations. RESULTS: The halothane dose was similar in all groups. Methoxsalen decreased 0- to 8-h trifluoroacetic acid (23 +/- 20 micromol vs 116 +/- 78 micromol) and bromide (17 +/- 11 micromol vs 53 +/- 49 micromol) excretion (P < 0.05), but not thereafter. Plasma F2-isoprostanes in controls were increased from 8.5 +/- 4.5 pg/ml to 12.5 +/- 5.0 pg/ml postoperatively (P < 0.05). Neither methoxsalen nor troleandomycin diminished reductive halothane metabolite or F2-isoprostane concentrations. CONCLUSIONS: These results provide the first evidence for halothane-dependent lipid peroxidation in humans. Methoxsalen effects on halothane oxidation confirm in vitro results and suggest limited CYP2A6 participation in vivo. CYP2A6-mediated, like CYP2E1-mediated human halothane oxidation, can be inhibited in vivo by mechanism-based CYP inhibitors. In contrast, clinical halothane reduction and lipid peroxidation were not amenable to suppression by CYP inhibitors.


Anesthetics, Inhalation/metabolism , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Halothane/metabolism , Lipid Peroxidation/drug effects , Mixed Function Oxygenases/metabolism , Adult , Aged , Anesthetics, Inhalation/pharmacokinetics , Bromides/urine , Chlorofluorocarbons/analysis , Cytochrome P-450 CYP2A6 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Dinoprost/blood , Enzyme Inhibitors/pharmacology , Female , Fluorides/urine , Halothane/analogs & derivatives , Halothane/analysis , Halothane/pharmacokinetics , Humans , Male , Methoxsalen/pharmacology , Middle Aged , Mixed Function Oxygenases/antagonists & inhibitors , Trifluoroacetic Acid/urine , Troleandomycin/pharmacology
4.
Anesthesiology ; 90(3): 766-71, 1999 Mar.
Article En | MEDLINE | ID: mdl-10078678

BACKGROUND: Some evidence suggests that isoflurane metabolism to trifluoroacetic acid and inorganic fluoride by human liver microsomes in vitro is catalyzed by cytochrome P450 2E1 (CYP2E1). This investigation tested the hypothesis that P450 2E1 predominantly catalyzes human isoflurane metabolism in vivo. Disulfiram, which is converted in vivo to a selective inhibitor of P450 2E1, was used as a metabolic probe for P450 2E1. METHODS: Twenty-two elective surgery patients who provided institutionally-approved written informed consent were randomized to receive disulfiram (500 mg orally, N = 12) or nothing (controls, N = 10) the evening before surgery. All patients received a standard isoflurane anesthetic (1.5% end-tidal in oxygen) for 8 hr. Urine and plasma trifluoroacetic acid and fluoride concentrations were quantitated in samples obtained for 4 days postoperatively. RESULTS: Patient groups were similar with respect to age, weight, gender, duration of surgery, blood loss, and delivered isoflurane dose, measured by cumulative end-tidal isoflurane concentrations (9.7-10.2 MAC-hr). Postoperative urine excretion of trifluoroacetic acid (days 1-4) and fluoride (days 1-3) was significantly (P<0.05) diminished in disulfiram-treated patients. Cumulative 0-96 hr excretion of trifluoroacetic acid and fluoride in disulfiram-treated patients was 34+/-72 and 270+/-70 micromoles (mean +/- SD), respectively, compared to 440+/-360 and 1500+/-800 micromoles in controls (P<0.05 for both). Disulfiram also abolished the rise in plasma metabolite concentrations. CONCLUSIONS: Disulfiram, a selective inhibitor of human hepatic P450 2E1, prevented 80-90% of isoflurane metabolism. These results suggest that P450 2E1 is the predominant P450 isoform responsible for human clinical isoflurane metabolism in vivo.


Anesthetics, Inhalation/administration & dosage , Cytochrome P-450 CYP2E1/metabolism , Disulfiram/administration & dosage , Enzyme Inhibitors/administration & dosage , Isoflurane/administration & dosage , Administration, Oral , Adult , Aged , Cytochrome P-450 CYP2E1 Inhibitors , Female , Humans , Male , Middle Aged , Trifluoroacetic Acid/blood , Trifluoroacetic Acid/urine
5.
J Chromatogr B Biomed Sci Appl ; 692(2): 413-8, 1997 May 09.
Article En | MEDLINE | ID: mdl-9188831

Halothane (CF3CHClBr), a widely used volatile anesthetic, undergoes extensive biotransformation in humans. Oxidative halothane metabolism yields the stable metabolites trifluoroacetic acid and bromide which can be detected in plasma and urine. To date, analytical methodologies have either required extensive sample preparation, or two separate analytical procedures to determine plasma and urine concentrations of these analytes. A rapid and sensitive method utilizing high-performance liquid chromatography-ion chromatography (HPLC-IC) with suppressed conductivity detection was developed for the simultaneous detection of both trifluoroacetic acid and bromide in plasma and urine. Sample preparation required only ultrafiltration. Standard curves were linear (r2> or =0.99) from 10 to 250 microM trifluoroacetic acid and 2 to 5000 microM bromide in plasma and 10 to 250 microM trifluoroacetic acid and 2 to 50 microM bromide in urine. The assay was applied to quantification of trifluoroacetic acid and bromide in plasma and urine of a patient undergoing halothane anesthesia.


Bromides/blood , Bromides/urine , Chromatography, High Pressure Liquid/methods , Halothane/pharmacokinetics , Trifluoroacetic Acid/blood , Trifluoroacetic Acid/urine , Humans , Ions , Sensitivity and Specificity
6.
Xenobiotica ; 27(1): 87-99, 1997 Jan.
Article En | MEDLINE | ID: mdl-9041681

1. Previous studies with the halothane analogue and chlorofluorocarbon replacement 2,2-dichloro-1,1,1-trifluoroethane (HCFC-123) have shown that there are concentration-dependent, sex-specific differences in the rate of uptake during inhalation exposure in rat. Since it is well established that there are sex-specific differences in the control of enzyme activity in drug metabolism, male and female rats were exposed by inhalation to halothane concentrations ranging from 500 to 4000 ppm. 2. A physiologically based pharmacokinetic model describing the concentration-dependent reduction in uptake and metabolism of halothane in male and female rats was developed. The in vivo metabolic rate constants obtained were: for male rats, Km = 0.4 mg litre-1 (2.03 mumol litre-1) and Vmaxc = 9.2 mg kg1 h-1 (46.6 mumol kg1 h-1); for female rats, Km = 0.4 mg litre-1 (2.03 mumol litre-1) and Vmaxc = 10.2 mg kg-1 h-1 (51.7 mumol kg-1 h-1). 3. An equation describing the concentration-dependent decrease of hepatic metabolism of halothane successfully simulated the gas-uptake data. Simulation of cumulative urinary excretion of the major metabolite, trifluoroacetic acid, required introduction of a proportionality constant to limit the extent of reduction of halothane metabolism to 20% of the amount of enzyme activity. Good simulation of urinary excretion data was achieved, which was interpreted to indicate that, when only 20% of the enzyme is inactivated, the rate of enzyme resynthesis was adequate to replenish enzyme activity within 24 h. 4. A rapidly reversible, non-biological inactivation mechanism called "physical toxicity' is discussed as a possible explanation of concentration-dependent gas uptake.


Halothane/metabolism , Halothane/pharmacokinetics , Animals , Chlorofluorocarbons/metabolism , Chlorofluorocarbons/pharmacokinetics , Female , Halothane/blood , Halothane/urine , Kinetics , Male , Models, Biological , Rats , Rats, Sprague-Dawley , Sex Factors , Tissue Distribution , Trifluoroacetic Acid/urine
7.
Lancet ; 347(9012): 1367-71, 1996 May 18.
Article En | MEDLINE | ID: mdl-8637342

BACKGROUND: Fulminant hepatic necrosis ("halothane hepatitis") is an unusual and often fatal complication of halothane anaesthesia. It is mediated by immune sensitisation in susceptible individuals to trifluoroacetylated liver protein neoantigens, formed by oxidative halothane metabolism. The seminal event in halothane hepatitis is hepatic metabolism, yet the enzyme responsible for oxidative halothane metabolism and trifluoroacetylated neoantigen formation remains unidentified. This investigation tested the hypothesis that cytochrome P450 2E1 (CYP2E1) is responsible for human halothane metabolism in vivo. METHODS: 20 elective surgical patients received either disulfiram (500 mg orally, n = 10) or nothing (controls, n = 10) the night before surgery. Disulfiram, converted in vivo to an effective inhibitor of P450 2E1, was used as a metabolic probe for P450 2E1. All patients received standard halothane anaesthesia (1.0% end-tidal, 3 h). Blood halothane and plasma and urine trifluoroacetic acid, bromide, and fluoride concentrations were measured for up to 96 h postoperatively. FINDINGS: Total halothane dose, measured by cumulative end-tidal (3.8 SE 0.1 minimum alveolar concentration hours) and blood halothane concentrations, was similar in the two groups. Plasma concentrations and urinary excretion of trifluoroacetic acid and bromide, indicative of oxidative and total (oxidative and reductive) halothane metabolism, respectively, were significantly diminished in disulfiram-treated patients. In control and disulfiram-treated patients cumulative 96 h postoperative trifluoroacetic acid excretion was 12,900 (SE 1700) and 2010 (440) mumol, respectively (p < 0.001) while that of bromide was 1720 (290) and 160 (70) mumol (p < 0.001). INTERPRETATION: The substantial attenuation of trifluoroacetic acid production by disulfiram after halothane anaesthesia suggests that P450 2E1 is a predominant enzyme responsible for human oxidative halothane metabolism. Inhibition of P450 2E1 by a single preoperative oral disulfiram dose greatly diminished production of the halothane metabolite responsible for the neoantigen formation that initiates halothane hepatitis. Single-dose disulfiram may provide effective prophylaxis against halothane hepatitis.


Cytochrome P-450 Enzyme Inhibitors , Disulfiram/pharmacology , Halothane/metabolism , Hepatic Encephalopathy/chemically induced , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Adult , Alanine Transaminase/blood , Cytochrome P-450 CYP2E1 , Cytochrome P-450 Enzyme System/metabolism , Disulfiram/therapeutic use , Halothane/adverse effects , Hepatic Encephalopathy/enzymology , Hepatic Encephalopathy/prevention & control , Humans , Male , Oxidoreductases, N-Demethylating/metabolism , Preoperative Care , Trifluoroacetic Acid/blood , Trifluoroacetic Acid/urine
8.
Drug Metab Dispos ; 23(8): 875-80, 1995 Aug.
Article En | MEDLINE | ID: mdl-7493556

Gas-uptake pharmacokinetics and metabolism of the chlorofluorocarbon replacement 2-chloro-1,1,1,2-tetrafluoroethane (HCFC-124) were investigated in rats, mice, and hamsters. Species differences in the rate of uptake of HCFC-124 and urinary excretion of trifluoroacetic acid were observed. In rats and mice, the uptake of HCFC-124 was described by both saturable and first-order components, whereas in the hamster only first-order uptake was observed. The in vivo metabolic rate constants obtained from computer simulation of the gas-uptake data were: for rats-KM = 1.2 mg liter-1 (8.79 mmol liter-1, Vmaxc = 0.35 +/- 0.01 mg kg-1 hr-1 (2.56 +/- 0.01 mmol kg-1 hr-1), and kfc = 1.25 +/- 0.01 hr-1 kg231; for mice-KM = 1.2 mg liter-1 (8.79 mmol liter-1), Vmaxc = 1.78 +/- 0.01 mg kg-1 hr-1 (13.0 +/- 0.007 mmol kg-1 hr-1), and kfc = 4.08 +/- 0.01 hr-1 kg-1; and for hamsters-kfc = 1.47 +/- 0.02 hr-1 kg-1. The production and excretion of trifluoroacetic acid, the major urinary metabolite of HCFC-124, were also simulated in rats and mice, but not in hamsters, by the physiologically based pharmacokinetic model when the in vivo metabolic rate constants obtained in the gas-uptake simulation studies were used. The blood:air partition coefficient of HCFC-124 in the hamster was lower than in the rat or mouse. A low blood:air partition coefficient may limit the pulmonary uptake of volatile chemicals.(ABSTRACT TRUNCATED AT 250 WORDS)


Chlorofluorocarbons, Methane/metabolism , Chlorofluorocarbons, Methane/pharmacokinetics , Animals , Body Weight/physiology , Chlorofluorocarbons, Ethane , Computer Simulation , Cricetinae , Male , Mesocricetus , Mice , Models, Biological , Rats , Rats, Sprague-Dawley , Regional Blood Flow/physiology , Species Specificity , Trifluoroacetic Acid/urine
9.
J Pharm Biomed Anal ; 12(12): 1489-93, 1994 Dec.
Article En | MEDLINE | ID: mdl-7696372

In this work, oxidative metabolism of the new propellant, 1,1,1,2-tetrafluoroethane to trifluoroacetic acid in man is shown to be minimal. Alternative propellants and refrigerants are under development to replace the currently used chlorofluorocarbons which lead to stratospheric ozone depletion. One potentially useful replacement is the hydrofluorocarbon, 1,1,1,2-tetrafluoroethane (HFA-134a). Before it can be used, however, particularly as a propellant in an aerosol pharmaceutical formulation whereby the compound is in effect dosed to people, it is important that the safety of this compound is established. As a part of this safety evaluation it is necessary to understand the metabolism of HFA-134a. In this work the production of the potential oxidative metabolite of HFA-134a, trifluoroacetic acid (TFA) has been studied in human urine following inhalation dosing with HFA-134a. The concentrations of TFA in urine have been measured using a highly sensitive 19F nuclear magnetic resonance procedure with a limit of detection of 10 ng ml-1 based on an acquisition time of only 2.25 h per sample. TFA is the only fluorinated species observed in the urine samples and only at very low levels, indicating that the oxidative route of metabolism can occur in vivo in man, but this metabolism is minimal in terms of percentage of administered dose.


Hydrocarbons, Fluorinated/metabolism , Trifluoroacetic Acid/urine , Adult , Humans , Hydrocarbons, Fluorinated/administration & dosage , Magnetic Resonance Spectroscopy/methods , Male , Middle Aged
10.
Toxicol Appl Pharmacol ; 129(1): 103-13, 1994 Nov.
Article En | MEDLINE | ID: mdl-7974482

2,2-Dichloro-1,1,1-trifluorethane (HCFC-123) is used industrially as a refrigerant, as a foam blowing agent, and as a solvent. It is also being considered as a replacement for halons and chlorinated fluorocarbons which have been banned by the Montreal Protocol because they deplete atmospheric ozone. Male Fischer 344 rats were exposed to 1.0, 0.1, and 0.01% HCFC-123 by inhalation. Parent compound was measured in blood, fat, and exhaled breath and trifluoroacetic acid (TFA) was measured in blood and urine. A physiologically based pharmacokinetic (PBPK) model was developed which included a gut compartment and a variable size fat compartment in addition to the standard flow-limited compartments. Compartment volumes and flows were chosen from the literature, partition coefficients were measured in the laboratory, and metabolic parameters were optimized from experimental data using model simulations. Laboratory experiments showed that the TFA blood concentration during the 1.0% exposure was more than 50% less than the TFA blood concentration during the 0.1% exposure. After cessation of the 4-hr exposure, TFA blood concentrations from the 1.0% exposure rebounded and peaked between 12 and 26 hr after the exposure at about the same concentration as the 0.1% peak. This rebound phenomenon suggested that it was not killing of the metabolic enzymes but substrate inhibition that made the TFA blood concentrations lower than expected. Substrate inhibition by halothane, a structural analog of HCFC-123, has been described in the literature. Only by including a term for substrate inhibition in the PBPK model could pharmacokinetic data for TFA in blood be simulated adequately. This combination of laboratory experimentation and PBPK modeling can be applied to relate the levels of parent and metabolite to toxic effects with some hope of elucidating the toxic species. This work is the first step toward developing models that can be used to predict the toxicokinetics of HCFC-123 in humans throughout various potential use scenarios.


Adipose Tissue/metabolism , Chlorofluorocarbons/pharmacokinetics , Trifluoroacetic Acid/blood , Adipose Tissue/drug effects , Administration, Inhalation , Animals , Blood Chemical Analysis , Breath Tests , Chlorofluorocarbons/administration & dosage , Chlorofluorocarbons/toxicity , Chlorofluorocarbons, Ethane , Chromatography, Gas , Dose-Response Relationship, Drug , Flame Ionization , Male , Models, Biological , Models, Theoretical , Rats , Rats, Inbred F344 , Trifluoroacetic Acid/urine
11.
Drug Metab Dispos ; 22(4): 511-7, 1994.
Article En | MEDLINE | ID: mdl-7956723

The in vivo metabolic rate constants for the metabolism of the chlorofluorocarbon replacement 2,2-dichloro-1,1,1-trifluoroethane (HCFC-123) were determined for both male and female rats with a physiologically based pharmacokinetic model. Uptake studies with 500-5,000 ppm HCFC-123 indicated that a single saturable component was involved in both sexes, and no significant differences were observed in in vivo metabolic rate constants between male and female rats. The in vivo metabolic rate constants obtained from computer simulation studies were: for male rats--KM = 1.2 mg liter-1 (7.85 mumol liter-1) and Vmaxc = 7.20 +/- 0.28 mg kg-1 hr-1 (47.1 +/- 1.83 mumol kg-1 hr-1); for female rats--KM = 1.2 mg liter-1 (7.85 mumol liter-1) and Vmaxc = 7.97 +/- 0.30 mg kg-1 hr-1 (52.1 +/- 1.96 mumol kg-1 hr-1). The physiologically based pharmacokinetic model failed to simulate the reduction in HCFC-123 uptake in female rats at 2,000-5,000 ppm. The production and excretion of trifluoroacetic acid, the major urinary metabolite of HCFC-123, was also predicted by the physiologically based pharmacokinetic model with in vivo metabolic rate constants obtained in the gas-uptake simulation studies. Diallyl sulfide, a selective, mechanism-based inhibitor of cytochrome P450 2E1, inhibited the metabolism of HCFC-123, as indicated by a decreased uptake of HCFC-123 and by a lowered urinary excretion of trifluoroacetic acid in diallyl sulfide-treated rats.


Allyl Compounds , Chlorofluorocarbons/pharmacokinetics , Administration, Oral , Animals , Biotransformation , Chlorofluorocarbons, Ethane , Computer Simulation , Cytochrome P-450 CYP2E1 , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/urine , Drug Interactions , Female , Gases/pharmacokinetics , Male , Models, Biological , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/urine , Rats , Rats, Sprague-Dawley , Sulfides/pharmacology , Trifluoroacetic Acid/urine
12.
Chem Res Toxicol ; 5(5): 720-5, 1992.
Article En | MEDLINE | ID: mdl-1446014

The hydrochlorofluorocarbons (HCFCs) 2,2-dichloro-1,1,1-trifluoroethane (HCFC-123) and 2-chloro-1,1,1,2-tetrafluoroethane (HCFC-124) and the hydrofluorocarbon (HFC) pentafluoroethane (HFC-125) are being developed as substitutes for chlorofluorocarbons that deplete stratospheric ozone. The structural similarity of these HCFCs and HFCs to halothane, which is hepatotoxic under certain circumstances, indicates that the metabolism and cellular interactions of HCFCs and HFCs must be explored. In a previous study [Harris et al. (1991) Proc. Natl. Acad. Sci. U.S.A. 88, 1407], similar patterns of trifluoroacetylated proteins (TFA-proteins) were detected by immunoblotting with anti-TFA-protein antibodies in livers of rats exposed to halothane or HCFC-123. The present study extends these results and demonstrates that in vivo TFA-protein formation resulting from a 6-h exposure to a 1% atmosphere of these compounds follows the trend: halothane approximately HCFC-123 much greater than HFC-124, greater than HFC-125. The calculated enthalpies of activation of halothane, HCFC-123, HCFC-124, and HFC-125 paralleled the observed rate of trifluoroacetic acid excretion in HCFC- or HFC-exposed rats. Exposure of rats to a range of HCFC-123 concentrations indicated that TFA-protein formation was saturated at an exposure concentration between 0.01% and 0.1% HCFC-123. Deuteration of HCFC-123 decreased TFA-protein formation in vivo. Urinary trifluoroacetic acid excretion by treated rats correlated with the levels of TFA-proteins found after each of these treatments. No TFA-proteins were detected in hepatic fractions from rats given 1,1,1,2-tetrafluoroethane (HFC-134a), which is not metabolized to a trifluoroacetyl halide.(ABSTRACT TRUNCATED AT 250 WORDS)


Chlorofluorocarbons, Methane/metabolism , Chlorofluorocarbons/metabolism , Fluorocarbons/metabolism , Microsomes, Liver/drug effects , Proteins/metabolism , Trifluoroacetic Acid/urine , Animals , Biotransformation , Chlorofluorocarbons/chemistry , Chlorofluorocarbons/toxicity , Chlorofluorocarbons, Ethane , Chlorofluorocarbons, Methane/chemistry , Chlorofluorocarbons, Methane/toxicity , Electrophoresis, Polyacrylamide Gel , Fluorocarbons/chemistry , Fluorocarbons/toxicity , Immunoblotting , Male , Microsomes, Liver/metabolism , Protein Binding , Rats , Rats, Inbred F344
13.
Biochem Pharmacol ; 42(11): 2229-38, 1991 Nov 06.
Article En | MEDLINE | ID: mdl-1958238

2,2,2-Trifluoroethanol (TFE) is a common industrial solvent and a known metabolite of the inhalation anesthetics fluroxene (2,2,2-trifluoroethyl vinyl ether) and halothane (2-bromo-2-chloro-1,1,1-trifluoroethane). The water-soluble metabolites of TFE were identified in the urine of Sprague-Dawley rats using 19F NMR spectroscopy. In rats dosed with 0.21 g TFE/kg body weight, approximately one-half of the administered TFE was excreted as the trifluoroethyl glucuronide. The remaining TFE was oxidized, primarily to trifluoroacetaldehyde hydrate, with a small percentage of the aldehyde oxidized further to trifluoroacetate. One additional fluorinated compound was found; after investigation, this was identified as a Schiff's base compound resulting from the addition of trifluoroacetaldehyde to urea. The time-dependent excretion of TFE metabolites was measured as a function of ethanol induction of hepatic enzymes. This study demonstrates the utility of 19F NMR for the analysis of drug metabolism in laboratory animals. In addition, the resistance of trifluoroacetaldehyde hydrate to further oxidation, coupled with its reactivity with common cellular amines, indicates the potential toxicity of this metabolite to mammalian tissues.


Trifluoroethanol/urine , Acetaldehyde/analogs & derivatives , Acetaldehyde/pharmacokinetics , Acetaldehyde/urine , Animals , Ethanol/pharmacology , Fluorine , Magnetic Resonance Spectroscopy/methods , Male , Rats , Rats, Inbred Strains , Time Factors , Trifluoroacetic Acid/pharmacokinetics , Trifluoroacetic Acid/urine , Trifluoroethanol/pharmacokinetics
14.
Anesth Analg ; 73(2): 180-5, 1991 Aug.
Article En | MEDLINE | ID: mdl-1854033

We examined the metabolism of desflurane in 13 healthy volunteers given 7.35 +/- 0.81 MAC-hours (mean +/- SD) of desflurane and 26 surgical patients given 3.08 +/- 1.84 MAC-hours (mean +/- SD). Markers of desflurane metabolism included fluoride ion measured via an ion-specific electrode, nonvolatile organic fluoride measured after sodium fusion of urine samples, and trifluoroacetic acid determined by a gas chromatographic-mass spectrometric method. In both volunteer and patient groups, postanesthesia serum fluoride ion concentrations did not differ from background fluoride ion concentrations. Similarly, postanesthesia urinary excretion of fluoride ion and organic fluoride in volunteers was comparable to preanesthesia excretion rates. However, small but significant levels of trifluoroacetic acid were found in both serum and urine from volunteers after exposure to desflurane. A peak serum concentration of 0.38 +/- 0.17 mumol/L of trifluoroacetic acid and a peak urinary excretion rate of 0.169 +/- 0.107 mumol/h were detected in volunteers at 24 h after desflurane exposure. Although these increases in trifluoroacetic acid after exposure to desflurane were statistically significant, they are approximately 10-fold less than levels seen after exposure to isoflurane. Thus, desflurane strongly resists biodegradation, but a small amount is metabolized in humans.


Anesthetics/metabolism , Fluorides/blood , Isoflurane/analogs & derivatives , Trifluoroacetic Acid/blood , Administration, Inhalation , Adult , Desflurane , Fluorides/urine , Gas Chromatography-Mass Spectrometry , Humans , Isoflurane/metabolism , Male , Middle Aged , Trifluoroacetic Acid/urine
15.
Drug Metab Dispos ; 19(2): 419-22, 1991.
Article En | MEDLINE | ID: mdl-1676647

To determine the urinary and biliary excretion of metabolites of halothane in dogs, 12 beagles were anesthetized with halothane either at 0.5 MAC (minimum alveolar concentration) for 1 hr or at 1.4 MAC for 4 hr. Urine and bile were then collected for 11 days following the anesthesia. The concentrations of inorganic fluoride in the urine and bile were measured with a fluoride electrode and an ion meter. The concentration of total fluoride containing organic fluoride also was measured in the same manner after conversion of the organic fluoride to an inorganic form by combustion. The concentration of trifluoroacetic acid (TFA) in the urine was measured by ion chromatography and that in the bile by gas chromatography. Over 80% of all the fluoride was excreted in the urine as organic fluoride in both groups. While the fraction of TFA in the organic fluoride in the bile was about 30% in both groups, that in the urine was 40% in the 0.5 MAC group and 65% in the 1.4 MAC group. Therefore, it was concluded that the organic fluoride compounds, the metabolites of halothane, and in particular TFA, were excreted mostly into the urine. The extent of metabolism of halothane decreased from 7.6% in the 0.5 MAC group to 4.9% in the 1.4 MAC group. The urinary excretion rate of TFA, however, was not affected by the concentration of inspired halothane.


Bile/metabolism , Halothane/metabolism , Animals , Biotransformation , Chromatography, Gas , Chromatography, Ion Exchange , Dogs , Female , Fluorides/metabolism , Halothane/urine , Trifluoroacetic Acid/urine
16.
Br J Anaesth ; 64(4): 469-73, 1990 Apr.
Article En | MEDLINE | ID: mdl-2334621

An attempt was made in children to identify a urinary halothane-cysteine conjugate which had been described previously in adult patients following administration of halothane. If this conjugate was found it would indicate that a reductive metabolite of halothane binds covalently with the sulphydryl-containing amino acid, cysteine, a reaction which could lead to hepatic injury. The potential halothane-cysteine conjugate, N-acetyl-S-(2-bromo-2-chloro-1,1-difluoroethyl)-L-cysteine (acetyl BCFEC), was prepared and the identity of the compound established using hydrogen-1 and carbon-13 NMR spectroscopy and methane chemical ionization mass spectrometry. A measurement technique for acetyl BCFEC was developed using HPLC with u.v. detection at 200 nm. In six children after halothane anaesthesia, one child being studied twice, urine was collected for up to 1 week and analysed for acetyl BCFEC. Little or no acetyl BCFEC was detected in any of the 43 urine samples tested, indicating that in children it is not a significant urinary metabolite of halothane.


Acetylcysteine/analogs & derivatives , Anesthesia, Inhalation , Cysteine/metabolism , Halothane/metabolism , Acetylcysteine/urine , Biotransformation , Child , Chromatography, High Pressure Liquid , Halothane/pharmacokinetics , Humans , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Trifluoroacetic Acid/urine
17.
Br J Anaesth ; 64(4): 474-81, 1990 Apr.
Article En | MEDLINE | ID: mdl-2334622

Halothane (1% v/v inspired) was administered for 60 min to six children of mean age 74 months (range 14-119 months). Uptake of halothane was measured from the difference in the concentration in inspired and expired gas and varied from 176 to 310 mg kg-1, depending on minute ventilation. After administration of halothane ceased, its elimination in expired gas was measured in four patients until the conclusion of anaesthesia; 32-37% of the absorbed halothane was expired 90 min after halothane administration ceased. Urinary excretion of trifluoroacetic acid, fluoride and bromide was measured for up to 1 week. Of the absorbed halothane, 11.4% (range 6.3-18.2%) was excreted in urine as trifluoroacetic acid and 0.37% (range 0.10-0.64%) as inorganic fluoride. The urinary half-life of trifluoracetic acid was 41.8 h (range 10.4-59.1 h). The quantitative and qualitative metabolism of halothane via the reductive and oxidative pathways in children are comparable to values found in adults. No differences in the metabolism of halothane by children were found which would explain the different incidence of halothane-associated hepatitis compared with adults.


Anesthesia, Inhalation , Halothane/pharmacokinetics , Biotransformation/physiology , Bromides/urine , Child , Child, Preschool , Creatinine/urine , Fatty Liver/metabolism , Fluorides/urine , Half-Life , Humans , Infant , Male , Respiration , Trifluoroacetic Acid/urine
20.
Arzneimittelforschung ; 35(9): 1447-51, 1985.
Article En | MEDLINE | ID: mdl-3002400

Halothane (H), 2-bromo-2-chloro-1,1,1-trifluoroethane, was metabolized to trifluoroacetic acid (TFAA) over a very long time (several days) in rats after a 1 h exposure to the inhalational anesthetic. H inhibited its own metabolism as long as anesthetically active concentrations existed in the serum and tissue. That fraction of H which was not exhaled rapidly by the lung after the anesthesia was metabolized mainly over the time range from 5-48 h: 68% of the total amount of TFAA eliminated in the urine during 144 h (about 4 mg) were found in this time period. Disulfiram (D) dose dependently inhibited the formation of TFAA from H in vivo and in vitro, in liver microsomes of phenobarbital treated rats. Diethyldithiocarbamate (DDTC) and carbon disulfide (CS2), two metabolites of D, given to rats immediately after the H-anesthesia also reduced the metabolic formation of TFAA. However, if DDTC and CS2 were given 24 h before the anesthesia they caused only a small decrease in serum TFAA concentration. This finding indicates that both metabolites of D have only short-lasting inhibitory effects. In contrast, the inhibition of the oxidative metabolism of H by D seems to persist over a long time, since a small but significant decrease in the serum TFAA concentration was found even if D was given 72 h before the H-anesthesia. It was concluded that in vivo CS2 is really the active inhibitor. The short-lasting inhibitory effect of DDTC may be explained by its fast metabolic transformation into CS2, whereas the long-lasting effect of D is caused by its delayed degradation into this metabolite.


Carbon Disulfide/pharmacology , Disulfiram/pharmacology , Ditiocarb/pharmacology , Fluoroacetates/metabolism , Halothane/metabolism , Thiocarbamates/pharmacology , Trifluoroacetic Acid/metabolism , Anesthesia, Inhalation , Animals , Kidney/metabolism , Liver/metabolism , Male , Rats , Rats, Inbred Strains , Trifluoroacetic Acid/blood , Trifluoroacetic Acid/urine
...