Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 62(2): 191-8, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11389877

ABSTRACT

Oxidative ring opening of troglitazone (TGZ)(1) a thiazolidine 2,4-dione derivative used for the treatment of type II diabetes mellitus, leads to the formation of a quinone metabolite. The formation of TGZ quinone was shown to be NADPH dependent and to require active microsomal enzymes. Quinone formation was not affected by co-incubation with catalase or sodium azide and was partially inhibited (25%) by superoxide dismutase (SOD). Kinetic analysis of TGZ quinone formation in human liver microsomes implied single enzyme involvement. CYP3A isoforms were characterized as the primary enzymes involved in quinone formation by several lines of evidence including: (a) troleandomycin and ketoconazole almost completely inhibited microsomal quinone formation when SOD was present, whereas other CYP inhibitors had minimal effects (<20%); (b) TGZ quinone formation was highly correlated with regard to both contents (r(2): 0.9374) and activities (r(2): 0.7951) of CYP3A4 in human liver microsomes (HLM); (c) baculovirus insect cell-expressed human CYP3A4 was able to catalyze TGZ quinone formation at a higher capacity (V(max)/K(m)) than other human CYPs with the relative contribution of CYP3A4 in HLM estimated to be 20-fold higher than that of other CYPs; (d) TGZ quinone formation was increased by 350% in liver microsomes from rats pretreated with dexamethasone (DEX); and (e) plasma concentrations of TGZ quinone were increased by 260-680% in rats pretreated with DEX. The chemical nature of the quinone metabolite suggests an atypical CYP reaction consistent with a one-electron oxidation mechanism where an intermediate phenoxy radical combines with ferryl oxygen to subsequently form the quinone metabolite.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Chromans/metabolism , Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/metabolism , Oxidoreductases, N-Demethylating/metabolism , Thiazoles/metabolism , Thiazolidinediones , Animals , Baculoviridae/genetics , Cells, Cultured , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP3A , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Insecta , Kinetics , Microsomes, Liver/enzymology , Mixed Function Oxygenases/metabolism , Oxidation-Reduction/drug effects , Rats , Troglitazone
2.
Drug Metab Dispos ; 27(8): 916-23, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10421619

ABSTRACT

Atorvastatin (AT) is a second-generation potent inhibitor of 3-hydroxy-3-methylglutaryl-CoA reductase, clinically approved for lowering plasma cholesterol. Using a mixture of [D(5)/D(0)] AT and/or [(14)C]AT, the metabolic fate and excretion of AT were examined in rats and dogs following single and multiple oral doses. Limited biliary recycling was examined in one dog after a single dose of AT. AT-derived metabolites in bile samples were identified by metabolite screening of the [D(5)/D(0)] AT molecular clusters using tandem mass spectrometry. Bile was a major route of [(14)C] drug-derived excretion, accounting for 73 and 33% of the oral dose in the rat and dog, respectively. The remaining radioactivity was recovered in the feces; only trace amounts were excreted in urine. Radioactive components identified in rat and dog bile were the para- and ortho-hydroxy metabolites, a glucuronide conjugate of ortho-hydroxy AT, and unchanged AT. Two minor radioactive components were identified as beta-oxidation products of AT with one confirmed as a beta-oxidized AT derivative. The reappearance of AT and major metabolites in bile from a dog administered a sample of its previously excreted bile indicated biliary recycling is an important component in AT metabolism. Multiple dose administration in rats did not alter biliary metabolic profiles. Rat and dog plasma profiles after multiple dose administration were similar and showed no additional metabolites not found in bile. Examination of rat and dog bile and plasma indicates that AT primarily undergoes oxidative metabolism.


Subject(s)
Heptanoic Acids/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Pyrroles/pharmacokinetics , Animals , Atorvastatin , Bile/metabolism , Biotransformation , Chromatography, High Pressure Liquid , Dogs , Female , Glucuronidase/metabolism , Heptanoic Acids/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Male , Pyrroles/administration & dosage , Rats , Species Specificity , Spectrometry, Mass, Fast Atom Bombardment
3.
J Pharmacol Exp Ther ; 288(2): 791-7, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9918590

ABSTRACT

Mifepristone (RU486), an 11beta-substituted nor-steroid containing a 17alpha-1-propynyl group used clinically as an antiprogestin agent for medical abortions, was demonstrated to be a selective mechanism-based inactivator of human cytochrome P-450-3A4 (CYP-3A4). The loss of testosterone 6beta-hydroxylation activity was time- and concentration-dependent as well as requiring metabolism of mifepristone in a purified CYP-3A4 reconstituted system. The inactivation exhibited pseudofirst-order kinetics. The values for KI and kinactivation were 4.7 microM and 0.089 min-1, respectively. The reduced-CO spectrum of CYP-3A4 was decreased by 76%, whereas approximately 81% of the activity was lost following incubation with mifepristone in the reconstituted system in the presence of NADPH. However, the Soret peak of the inactivated CYP-3A4 was slightly increased. High-performance liquid chromatography analysis of the incubation mixture showed that the peak containing the heme dissociated from the inactivated CYP3A4 was almost identical with that seen for the -NADPH control. Covalent binding of [3H]mifepristone to apoCYP3A4 was demonstrated by SDS-PAGE and high-pressure liquid chromatography analyses of the reconstituted system containing CYP-3A4, NADPH-CYP reductase, cytochrome b5 and lipids in the presence of NADPH. The stoichiometry was determined to be approximately 1 mol of mifepristone bound per 1 mol of CYP-3A4 inactivated. Therefore, the mechanism of inactivation of CYP-3A4 by mifepristone involves irreversible modification of the apoprotein at the enzyme active site instead of being the result of heme adduct formation or heme fragmentation. Mifepristone exhibits selectivity for CYP-3A4 as evidenced by the fact that it did not show mechanism-based inactivation of CYPs 1A, 2B, 2D6, and 2E1, although a competitive inhibition of CYP 2B1 and 2D6 was observed.


PIP: This study demonstrates mifepristone (RU-486) as a potent and selective mechanism-based inactivator of cytochrome P-450-3A4 (CYP-3A4) via irreversible modification of the apoprotein. The results of this clinical research indicate that loss of testosterone 6-beta-hydroxylation activity was time- and concentration-dependent, as well as requiring metabolism of mifepristone in purified CYP-3A4 reconstituted system. Inactivation using several different concentrations of mifepristone exhibited pseudo-first-order kinetics. Reduced-CO difference spectrum of CYP-3A4 decreased by 76%, whereas approximately 81% of CYP-3A4 activity was lost following incubation with mifepristone, indicating the occurrence of N-heme adduct formation detected by HPLC and UV-visible spectroscopy. The peak containing the heme dissociated from the mifepristone-inactivated CYP-3A4 was almost identical with that of the -NADPH control when the incubation mixtures were analyzed by HPLC. 3H-mifepristone proved to be covalently bound to the apoCYP-3A4 by HPLC and SDC-PAGE. The stoichiometry for the binding of the mifepristone was determined to be 1.02 +or- 0.15, approximately 1 mol of mifepristone bound per mole of inactivated CYP-3A4. In summary, mifepristone has been shown to be a potent mechanism-based inactivator of human CYP-3A4. The mechanism of the inactivation showed to involve irreversible modification of the apoprotein at the enzyme active site instead of heme adduct formation or heme fragmentation.


Subject(s)
Abortifacient Agents, Steroidal/pharmacology , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Mifepristone/pharmacology , Mixed Function Oxygenases/drug effects , Mixed Function Oxygenases/metabolism , Abortifacient Agents, Steroidal/metabolism , Animals , Apoenzymes/drug effects , Apoenzymes/metabolism , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , Heme/metabolism , Humans , Kinetics , Male , Mifepristone/metabolism , Mixed Function Oxygenases/antagonists & inhibitors , NADP/metabolism , Rats , Rats, Inbred F344 , Sodium Dodecyl Sulfate , Spectrum Analysis/methods , Testosterone/metabolism
4.
Br J Clin Pharmacol ; 46(3): 221-8, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9764962

ABSTRACT

AIMS: To study the potential utility of caffeine based probes of CYP1A2 enzyme activity in predicting the pharmokinetics of tacrine in patients with Alzheimer's disease. METHODS: The pharmokinetics of a single 40 mg oral dose of tacrine were measured in 19 patients with Alzheimer's disease. Each patient also received 2 mg kg(-1) [13C-3-methyl] caffeine orally and had breath and urine samples collected. RESULTS: Tacrine oral clearance (CL F(-1) kg(-1)), which varied 15-fold among the patients, correlated significantly with the 2 h total production of 13CO2 in breath (r=0.56, P=0.01), and with each of two commonly used urinary caffeine metabolite ratios: the 'paraxanthine/caffeine ratio' (1,7X + 1, 7U)/1,3,7X) (r=0.76, P=0.0002) and the 'caffeine metabolic ratio' (AFMU + 1X + 1U)/1, 7U)(r=0.76, P=0.0001). CONCLUSIONS: These observations support a central role for CYP1A2 in the in vivo disposition of tacrine and the potential for drug interactions when tacrine treated patients receive known inducers or inhibitors of this enzyme. The magnitude of the correlations we observed, however, are probably not sufficient to be clinically useful in individualizing tacrine therapy.


Subject(s)
Alzheimer Disease/enzymology , Caffeine/pharmacokinetics , Cholinesterase Inhibitors/pharmacokinetics , Cytochrome P-450 CYP1A2/metabolism , Phosphodiesterase Inhibitors/pharmacokinetics , Tacrine/pharmacokinetics , Administration, Oral , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/urine , Caffeine/urine , Cholinesterase Inhibitors/adverse effects , Female , Humans , Male , Middle Aged , Phosphodiesterase Inhibitors/urine , Predictive Value of Tests , Tacrine/adverse effects
5.
Drug Metab Dispos ; 26(8): 755-63, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9698289

ABSTRACT

Atorvastatin, [(R-(R,R)]-2-(4-fluorophenyl)-beta, delta-dihydroxy-5-(1-methylethyl)-3-phenyl-4-[(phenyl-amino)carbonyl] -1H-pyrrole-1-heptanoic acid calcium salt (CI-981, AT), is a second generation 3-hydroxy-3-methylglutaryl-CoA reductase inhibitor approved for clinical use as a cholesterol lowering agent. The disposition and metabolism of AT, including potential CYP450 induction, was investigated in mice administered an oral dose of [14C]AT (free acid) on study days 1 and 14. Peak plasma radioactivity concentrations occurred 1 hr postdose after both single- and multiple-dose administration and declined rapidly thereafter. Total plasma radioactivity levels in mice receiving the multiple dose were approximately 50% of levels observed after single-dose administration. Plasma metabolic profiles, which provided evidence of extensive metabolism, remained similar. Feces was the major route of AT-derived radioactivity elimination. Fecal profiles showed extensive metabolism with qualitatively similar profiles after single- and multiple-dose administration; however, quantitative differences were apparent. Metabolites identified in plasma and feces include hydroxylated, beta-oxidized, and unsaturated derivatives of AT. Most metabolites had undergone beta-oxidation. In mice receiving multiple 1 mg/kg doses of AT, no effect on spectral P450 concentration was found, and only a minor increase was observed at the 200 mg/kg dose level. Catalytic activities of CYP4501A, -2B, and -3A were not significantly affected; CYP4A activity decreased in a dose-dependent manner. Administration of multiple doses resulted in lower systemic plasma levels of total AT-derived radioactivity not readily explained by these studies. In mice, the majority of metabolites are formed primarily through the beta-oxidation pathway.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Heptanoic Acids/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Pyrroles/pharmacokinetics , Administration, Oral , Animals , Anticholesteremic Agents/blood , Anticholesteremic Agents/metabolism , Atorvastatin , Carbon Radioisotopes , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Drug Administration Schedule , Enzyme Induction , Feces/chemistry , Heptanoic Acids/blood , Heptanoic Acids/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Liver/drug effects , Liver/enzymology , Male , Mice , Mice, Inbred Strains , Pyrroles/blood , Pyrroles/metabolism
6.
Chem Res Toxicol ; 11(4): 252-9, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9548795

ABSTRACT

Grapefruit juice has been found to significantly increase oral bioavailability of several drugs metabolized by cytochrome P450 3A4 (P450 3A4) through inhibiting the enzymatic activity and decreasing the content of intestinal P450 3A4. HPLC/MS/MS and HPLC/UV analyses of ethyl acetate extracts from grapefruit juice revealed the presence of several furanocoumarins of which bergamottin (BG) is the major one. BG was shown to inactivate P450 3A4 in a reconstituted system consisting of purified P450 3A4, NADPH-cytochrome P450 reductase, cytochrome b5, and phospholipids. Inactivation was time- and concentration-dependent and required metabolism of BG. The loss of catalytic activity exhibited pseudo-first-order kinetics. The values of kinactivation and KI calculated from the inactivation studies were 0.3 min-1 and 7.7 microM, respectively. While approximately 70% of the erythromycin N-demethylation activity was lost during incubation with BG in the reconstituted system, P450 3A4 retained more than 90% of the heme as determined either by UV-visible spectroscopy or by HPLC. However, approximately 50% of the apoP450 in the BG-inactivated P450 3A4 incubation mixture could not be recovered from a reverse-phase HPLC column when compared with the -NADPH control. The mechanism of the inactivation appears to involve modification of the apoP450 in the active site of the enzyme instead of heme adduct formation or heme fragmentation. These results indicate that BG, the primary furanocoumarin extracted from grapefruit juice, is a mechanism-based inactivator of P450 3A4. BG was also found to inhibit the activities of P450s 1A2, 2A6, 2C9, 2C19, 2D6, 2E1, and 3A4 in human liver microsomes.


Subject(s)
Beverages , Citrus , Cytochrome P-450 Enzyme Inhibitors , Furocoumarins/pharmacology , Mixed Function Oxygenases/antagonists & inhibitors , Cytochrome P-450 CYP3A , Furocoumarins/analysis , Humans
7.
Biochem Pharmacol ; 54(3): 425-7, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9278102

ABSTRACT

The effect of multiple-dose tacrine (THA) administration at 2 and 20 mg/kg (single oral doses for 2 weeks) on cytochrome P450 (CYP) was examined in male and female Wistar rats. Changes in CYP were determined by measuring total spectral CYP, the rates of ethoxy- and pentoxyresorufin dealkylations, and the protein expression of several CYPs by western blot analysis of liver microsomes. Animals treated with beta-naphthoflavone or phenobarbital were employed as positive controls. No physiological or metabolic changes were observed in male or female rats treated with 2 mg/kg THA for 2 weeks. Male and female animals treated with 20 mg/kg THA for 2 weeks demonstrated increased CYP1A activity (increased ethoxyresorufin deethylase activity) and increased expression of CYP1A1 with only minor increases in CYP1A2 expression. Compared with the effects of beta-naphthoflavone induction of CYP1A, the induction observed with THA at 20 mg/kg was considered minor.


Subject(s)
Cholinesterase Inhibitors/administration & dosage , Cytochrome P-450 Enzyme System/biosynthesis , Isoenzymes/biosynthesis , Microsomes, Liver/drug effects , Tacrine/administration & dosage , Animals , Benzoflavones/pharmacology , Body Weight , Enzyme Induction , Female , Male , Microsomes, Liver/enzymology , Phenobarbital/pharmacology , Rats , Rats, Wistar
8.
Drug Metab Dispos ; 25(5): 590-7, 1997 May.
Article in English | MEDLINE | ID: mdl-9152598

ABSTRACT

The metabolic fate of tacrine [1,2,3,4-tetrahydro-9-acridinamine monohydrochloride monohydrate (THA)] was examined in rats, dogs, and humans. After administration of single oral doses of [14C]THA to rats, dogs, and humans, drug-derived material was well absorbed, with urinary excretion being the predominant route of radiolabel elimination. Metabolic profiling of plasma and urine from rats, dogs, and humans showed THA to be extensively metabolized with marked species differences in quantitative amounts of metabolites observed. Plasma profiles were similar to respective urinary profiles in all three species. Present in profiles of urine from rats were 1-hydroxy (OH)-THA (major), 2-OH-THA, and 4-OHA-THA, and unchanged THA. Also observed were trace amounts of more polar metabolites, presumably arising from sequential metabolism. Metabolic profiling of dog urine also showed 1-OH-THA to be the major metabolite, with trace amounts of the 2-OHA-THA and 4-OH-THA regioisomers and THA excreted. In dog urine, more of the radioactivity was associated with polar metabolites, including 1,3-dihydroxy-THA and a dihydrodiol metabolite. Human urinary metabolic profiles were more similar to that in dogs than in rats, with no single metabolite constituting > 10% of urinary radioactivity. Present in human urine were phenol glucuronide metabolites, of which 7-OH-THA was identified as an aglycone. Relevance of the marked quantitative differences in THA metabolism between rats, dogs, and humans to species differences in THA hepatotoxic potential remains to be established.


Subject(s)
Cholinesterase Inhibitors/urine , Nootropic Agents/urine , Tacrine/urine , Animals , Cholinesterase Inhibitors/pharmacokinetics , Chromatography, High Pressure Liquid , Dogs , Female , Humans , Magnetic Resonance Spectroscopy , Male , Nootropic Agents/pharmacokinetics , Rats , Rats, Wistar , Species Specificity , Tacrine/pharmacokinetics
9.
Drug Metab Dispos ; 25(1): 123-30, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9010639

ABSTRACT

The metabolism of CI-976, a potent inhibitor of liver and intestinal acyl coenzyme A:cholesterol acyltransferase, was investigated in isolated rat hepatocytes and Wistar rats after oral administration. The major metabolite observed both in vitro and in vivo was identified as the 6-carbon, chain-shortened 5,5-dimethyl-6-oxo-[(2,4,6-trimethoxyphenyl)amino]hexanoic acid (M-4). M-4 was determined to be formed from the omega-carboxylic acid 11,11-dimethyl-12-oxo-12-[(2,4,6-trimethoxyphenyl)amino]dodecanoic acid (M-1) via the 2- and 4-carbon, chain-shortened intermediate metabolites [9,9-dimethyl-10-oxo-10-[(2,4,6-trimethoxyphenyl)amino]decanoic acid (M-2) and 7,7-dimethyl-8-oxo-8-[(2,4,6-trimethoxyphenyl)amino]octanoic acid (M-3)], respectively. M-1 was, in turn, determined to be derived from omega-hydroxy CI-976. A minor metabolite, identified in vitro and in vivo, was a novel 5-carbon, chain-shortened derivative, 6,6-dimethyl-7-oxo-7-[(2,4,6-trimethoxyphenyl)amino]heptanoic acid (M-5). M-5 was shown not to be formed from either M-1 or the omega-hydroxy derivative. Separate incubation of CI-976 (omega-oxidation and beta-oxidation pathways) and M-1 (beta-oxidation only) indicated a potential gender difference in the omega-oxidation of CI-976. Both the omega-oxidation and beta-oxidation pathways were enhanced by clofibrate and phenobarbital induction, and CI-976 metabolism was completely inhibited when coincubated with SKF525A pointing to cytochrome P450-mediated metabolism, presumably CYP4A. Etomoxir and L-carnitine had minor effects on the beta-oxidation of M-1, indicating beta-oxidation occurs predominately within peroxisomes.


Subject(s)
Anilides/pharmacokinetics , Sterol O-Acyltransferase/antagonists & inhibitors , Anilides/metabolism , Anilides/urine , Animals , Cells, Cultured , Clofibrate/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Enzyme Induction , Feces/chemistry , Female , Liver/enzymology , Male , Phenobarbital/pharmacology , Rats , Rats, Wistar
10.
Xenobiotica ; 26(9): 935-46, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8893040

ABSTRACT

1. The disposition of tacrine 1,2,3,4-tetrahydro-9-acridinamine monohydrochloride monohydrate (THA, Cognex), was studied using livers obtained from control, phenobarbital (PB), isosafrole (ISO), and 3-methycholanthrene (3-MC) treated rats. 2. Pretreatment of rats with PB, ISO, and 3-MC reduced AUC(10-120 min) of THA in liver perfusates by 28, 32, and 86% respectively. 3. Elimination of [14C]-THA-derived radioactivity into bile was 7.6 +/- 1.2%, 11.7 +/- 2.9%, 14.8 +/- 2.0%, and 46.3 +/- 9.7% (mean +/- SD) of the infusion dose for control PB, ISO, and 3-MC pretreated isolated perfused rat livers, respectively. 4. In perfusion experiments using 3-MC pretreated livers, a marked increase in irreversible protein binding of 3-, 7-, and 8-fold was observed to microsomal, cytosolic and total liver proteins, respectively, compared to control. Only a slight effect was observed on protein binding in perfusion experiments using PB and ISO pretreated animals. 5. Co-incubations of [14C]-THA with the metabolic inhibitors enoxacin, ethimizol, and furafylline in hepatocyte preparations obtained from 3-MC pretreated rats markedly inhibited THA-derived irreversible protein binding. Furafylline, a specific inhibitor of cytochrome P4501A2, had the greatest inhibitory effect (approximately 70%). 6. These results are consistent with a major role of cytochrome P4501A in the metabolism and irreversible protein binding of THA in rat liver and demonstrate the utility of isolated liver perfusion and hepatocyte models for examining the effect of metabolic modulators.


Subject(s)
Cholinesterase Inhibitors/metabolism , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Tacrine/metabolism , Animals , Chromatography, High Pressure Liquid , Enoxacin/pharmacology , Enzyme Induction , Etimizol/pharmacology , Glucuronidase/metabolism , Glutathione/pharmacology , Liver/drug effects , Liver/enzymology , Male , Methylcholanthrene/pharmacology , Rats , Rats, Wistar , Theophylline/analogs & derivatives , Theophylline/pharmacology
11.
J Med Chem ; 39(15): 3014-8, 1996 Jul 19.
Article in English | MEDLINE | ID: mdl-8709135

ABSTRACT

Discrepancies in urinary metabolic profiles in rats administered tacrine (1) suggested the presence of an unidentified metabolite of 1. Chromatographic methods were developed that allowed isolation of a metabolite fraction containing both 1-hydroxytacrine (2) and an unknown metabolite from rat urine. Mass spectral analysis indicated this metabolite to be a monohydroxylated derivative, which upon two dimensional COSY NMR analysis could be assigned as 3-hydroxytacrine (4). This structural assignment was confirmed by independent synthesis of 4. Compound 4 was also identified as a human urinary metabolite of 1. Biologically, 4 was found to have in vitro human red blood cell acetylcholinesterase inhibitory activity similar to that of 2 and 4-hydroxytacrine (5) and approximately 8-fold less than that of 1. These results underscore the need to conduct rigorous structural identification studies, especially in cases where isomeric metabolites are possible, in assessing the accuracy of chromatographic profiling techniques.


Subject(s)
Cholinesterase Inhibitors/urine , Tacrine/analogs & derivatives , Tacrine/urine , Acetylcholinesterase/blood , Animals , Chromatography, High Pressure Liquid , Erythrocytes/enzymology , Humans , Hydroxylation , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Molecular Structure , Rats , Tacrine/chemistry
12.
Hepatology ; 23(6): 1429-35, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8675160

ABSTRACT

Therapy with tacrine, a promising new treatment for Alzheimer's disease, must be discontinued in up to 15% of patients because of hepatocellular toxicity. Recent studies using human liver microsomes have suggested that a single liver enzyme, cytochrome P450 1A2 (CYP1A2), catalyzes the major route of metabolism and elimination of tacrine, and also catalyzes the pathway(s) involved in the generation of reactive metabolites capable of covalent protein binding and cytotoxicity. Because CYP1A2 activity has been shown to vary up to 60-fold among patients, we proposed that a convenient measure of CYP1A2 activity, the [(13)C 3-methyl] caffeine breath test (CBT), might be clinically useful in identifying patients most susceptible to tacrine liver toxicity. To test this hypothesis, we administered the CBT to 37 patients with Alzheimer's disease before they began treatment with tacrine. Twenty patients received 2 mg/kg of [(13)C 3-methyl] caffeine. The remaining 17 patients received the commercially available CBT kit, which employs a constant 200-mg dose. The activities of two other major drug-metabolizing enzymes (cytochrome P450 3A4 and 2D6 [CYP3A4 and CYP2D6]) were also measured in these 17 patients. We found that the results obtained from the CBT protocol did not predict the peak serum alanine transaminase (ALT) observed in the patients. The measured CYP3A4 and CYP2D6 activities also failed to predict the susceptible patients. However, the result of the standardized-dose CBT correlated well with the logarithm of the steady-state plasma tacrine level obtained in 10 patients (R(2) = .69, P = .003). We conclude that the CBT will not be clinically useful in determining the subset of patients most susceptible to tacrine hepatotoxicity. However, the correlation we observed between CBT results and tacrine blood levels is the first evidence supporting a critical role for CYP1A2 activity in the disposition of the drug in vivo.


Subject(s)
Caffeine/pharmacokinetics , Cholinesterase Inhibitors/adverse effects , Liver/drug effects , Liver/metabolism , Tacrine/adverse effects , Aged , Aged, 80 and over , Alanine Transaminase/blood , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Breath Tests , Cholinesterase Inhibitors/blood , Cytochrome P-450 CYP1A2 , Cytochrome P-450 Enzyme System/metabolism , Female , Humans , Male , Middle Aged , Oxidoreductases/metabolism , Tacrine/blood , Tacrine/metabolism
13.
Drug Metab Dispos ; 24(6): 628-33, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8781777

ABSTRACT

Tacrine [1,2,3,4-tetrahydro-9-acridinamine monohydrochloride monohydrate (THA), Cognex] is a potent acetylcholinesterase inhibitor recently approved for treatment of mild-to-moderate Alzheimer's disease. The potential for THA and/or a metabolite of THA to accumulate in brain tissue was investigated by autoradiographic and metabolic profiling techniques in rats given single and multiple doses of [14C]THA. In addition, the brain-to-plasma distribution time course of orally administered 1-hydroxy-THA (1-OH-THA, 24 mg/kg), a primary rat metabolite with anticholinesterase activity, was also examined. Results from a 16 mg/kg single-dose study showed THA to cross the blood-brain barrier readily and concentrate in brain tissue, approximately 5-fold compared with plasma. The metabolite 1-OH-THA was found in much lower amounts relative to THA and when given separately at a similar dose the levels in brain tissue were comparable with plasma concentrations. After multiple-dose administration, THA concentrations in brain tissue were approximately 3-fold higher than those achieved after a single oral dose. However, concentration of 1-OH-THA metabolite increased only 50%. These data suggest a marked difference between the ability of THA and 1-OH-THA to accumulate in brain tissue and may reflect differences in lipophilicity as estimated by calculated log p values. The relevance of THA accumulation in brain tissue to delays observed in THA clinical management of Alzheimer's disease remains to be established.


Subject(s)
Brain/metabolism , Cholinesterase Inhibitors/pharmacokinetics , Tacrine/pharmacokinetics , Animals , Autoradiography , Cholinesterase Inhibitors/blood , Male , Rats , Rats, Wistar , Tacrine/analogs & derivatives , Tacrine/blood , Tissue Distribution/drug effects
14.
Xenobiotica ; 25(1): 103-16, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7604602

ABSTRACT

1. The metabolite profile of tacrine (1,2,3,4-tetrahydro-9-amino acridine) was similar in hepatic microsomes from man, rat, dog, rabbit, mouse and hamster. Major metabolites were 1-, 2-, 4- and 7-OH tacrine. Only quantitative differences in metabolite profile were evident between species. 2. Bioactivation to protein-reactive metabolite(s) was seen in microsomes from all species. 3. 7-Methyl tacrine was found to undergo significantly less bioactivation than either 7-OH tacrine or tacrine itself. 4. In the presence of hepatic microsomes and thiol-containing agents protein-reactive metabolite formation was significantly reduced. With mercaptoethanol present a stable thioether adduct was generated from both tacrine and 7-OH tacrine. 5. Analysis of the thioether adduct by mass spectrometry yielded a molecular ion of m/z 290 consistent with the presence of a covalent adduct of 7-OH tacrine complexed in a 1:1 molar ratio with mercaptoethanol. 6. We have therefore provided further evidence for a two-step mechanism in the bioactivation of tacrine involving an initial 7-hydroxylation followed by a postulated 2-electron oxidation to yield a reactive quinone methide. This mechanism of bioactivation appears to be identical in human and animal hepatic microsomes.


Subject(s)
Cholinesterase Inhibitors/metabolism , Microsomes, Liver/metabolism , Tacrine/metabolism , Amino Acids/pharmacology , Animals , Biotransformation , Chromatography, High Pressure Liquid , Cricetinae , Dogs , Humans , Hydroxylation/drug effects , Mass Spectrometry , Mice , NADP/metabolism , Protein Binding , Rabbits , Rats , Species Specificity , Sulfhydryl Compounds/pharmacology
15.
Xenobiotica ; 24(11): 1107-17, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7701852

ABSTRACT

1. Incubations of tacrine (1,2,3,4-tetrahydro-9-acridinamine monohydrochloride monohydrate, THA) with a primary suspension of rat hepatocytes for 2 min resulted in formation of the 1-hydroxy derivative as the major metabolite with smaller amounts of the 2- and 4-hydroxy metabolites. 2. Apparent Vmax and Km for THA metabolism were 12.4 +/- 3.3 nmol/min/g liver and 0.98 +/- 0.34 microM respectively. 3. Incubations of THA for longer time-periods (> 10 min) resulted in irreversible binding of THA-derived radioactivity to hepatocellular protein. The apparent maximal rate of irreversible binding (Bmax) was 76.7 +/- 30.5 pmol equivalents bound/h/mg cell protein, whereas the apparent Kb for binding was 2.8 +/- 1.4 microM. 4. The kinetic parameters, Vmax and Km, were used to predict steady-state extraction ratios (ERSS) for various THA input concentrations (Cin) in single-pass perfused rat liver. At low input concentrations (0.72-0.85 microM; Cin < Km), ERSS of THA was approximately 1. For higher Cin (14.05, 20.72, 20.88 microM; Cin >> Km), the calculated ERSS was markedly decreased with 0.300, 0.296 and 0.261, respectively. 5. The intrinsic clearance of THA (Cli) estimated from in vitro hepatocyte data was 6.7 ml/min/g liver while the apparent oral THA clearance (Cloral) calculated from in vivo rat data was 6.6 ml/min/g liver.


Subject(s)
Liver/metabolism , Tacrine/metabolism , Animals , Carbon Radioisotopes , In Vitro Techniques , Liver/chemistry , Liver/cytology , Male , Perfusion/methods , Prognosis , Rats , Rats, Wistar , Tacrine/administration & dosage , Tacrine/analysis
16.
Drug Metab Dispos ; 22(5): 719-24, 1994.
Article in English | MEDLINE | ID: mdl-7835223

ABSTRACT

An enantiospecific method was developed for assessing the stereochemistry of tacrine (9-amino-1,2,3,4-tetrahydroacridine monohydrochloride monohydrate; THA) metabolism to 1-hydroxytacrine (1-OH-THA) in humans and rats. In addition, limited metabolic studies with human liver microsomal preparations were conducted, and the stereochemistry of rac-1-OH-THA disposition was also examined. The analytical method incorporates an achiral normal phase separation and isolation of 1-OH-THA, followed by a chromatographic step using chiral normal-phase chromatography to resolve the enantiomers of 1-OH-THA. The achiral method was applied to quantitation of total 1-OH-THA in human urine specimens collected for 24 hr following administration of a single 40 mg oral dose of tacrine to 15 healthy elderly volunteers. Total 1-OH-THA accounted for approximately 5% of the administered dose. THA and 2-OH-THA were also quantitated and found to comprise < 1% and approximately 2% of the administered dose, respectively. 4-OH-THA was not detectable. The dextrorotatory (+)-isomer comprised approximately 94% of the 1-OH-THA recovered in urine. In vitro studies utilizing human liver microsomes found enantioselective formation of the (+)-isomer (approximately 90%), whereas incubations with rac-1-OH-THA showed residual substrate to be racemic. The method was also applied to determination of the enantiomeric composition of 1-OH-THA in the urine of rats given a single oral 16 mg/kg dose of THA. The percentage of 1-OH-THA excreted in urine as the (+)-isomer was 94%.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Tacrine/pharmacokinetics , Aged , Animals , Cholinesterase Inhibitors/urine , Chromatography, High Pressure Liquid , Female , Glucuronates/metabolism , Humans , Hydroxylation , Male , Microsomes, Liver/metabolism , Middle Aged , Rats , Stereoisomerism , Tacrine/analogs & derivatives , Tacrine/urine
17.
Br J Clin Pharmacol ; 38(1): 15-22, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7946932

ABSTRACT

1. Tacrine (1,2,3,4-tetrahydro-9-aminoacridine-hydrochloride: THA) underwent metabolism in vitro by a panel (n = 12) of human liver microsomes genotyped for CYP2D6, in the presence of NADPH, to both protein-reactive and stable metabolites. 2. There was considerable variation in the extent of THA metabolism amongst human livers. Protein-reactive metabolite formation showed a 10-fold variation (0.6 +/- 0.1%-5.2 +/- 0.8% of incubated radioactivity mg-1 protein) whilst stable metabolites showed a 3-fold variation (24.3 +/- 1.7%-78.6 +/- 2.6% of incubated radioactivity). 3. Using cytochrome P450 isoform specific inhibitors CYP1A2 was identified as the major enzyme involved in all routes of THA metabolism. 4. There was a high correlation between aromatic and alicyclic hydroxylation (r = 0.92, P < 0.0001) consistent with these biotransformations being catalysed by the same enzymes. 5. Enoxacin (ENOX), cimetidine (CIM) and chloroquine (CQ) inhibited THA metabolism by a preferential decrease in the bioactivation to protein-reactive, and hence potentially toxic, species. The inhibitory potency of ENOX and CIM was increased significantly upon pre-incubation with microsomes and NADPH. 6. Covalent binding correlated with 7-OH-THA formation before (r = 0.792, P < 0.0001) and after (r = 0.73, P < 0.0001) inhibition by CIM, consistent with a two-step mechanism in the formation of protein-reactive metabolite(s) via a 7-OH intermediate. 7. The use of enzyme inhibitors may provide a useful tool for examining the relationship between the metabolism and toxicity of THA in vivo.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Microsomes, Liver/enzymology , Tacrine/metabolism , Adult , C-Reactive Protein/biosynthesis , Child , Chloroquine/pharmacology , Cimetidine/pharmacology , Cytochrome P-450 Enzyme System/genetics , Enoxacin/pharmacology , Female , Humans , Hydroxylation , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Male , Microsomes, Liver/drug effects , Middle Aged , NADP/metabolism , Tacrine/pharmacokinetics
18.
Drug Metab Dispos ; 21(6): 1112-8, 1993.
Article in English | MEDLINE | ID: mdl-7905392

ABSTRACT

2,2-Dimethyl-N-(2,4,6-trimethoxyphenyl)dodecanamide, CI-976, is a newly developed fatty acid anilide being evaluated as a plasma lipid regulator and antiatherosclerotic agent. Disposition studies with CI-976 were conducted in rats and monkeys. In rats, CI-976 (suspension, 50 mg/kg) was approximately 30% bioavailable, with 46% of the administered radioactivity recovered in urine suggesting presystemic metabolism. The intravenous elimination half-life of CI-976 in rats was approximately 8 hr. Radioactivity derived from [14C]CI-976 was almost completely recovered in rats after either oral or intravenous administration. In bile duct-cannulated rats receiving either an oral or intravenous dose, biliary excretion was the major route of drug-derived radioactivity elimination. There was no evidence for unchanged drug in urine or bile. In monkeys, 13% urinary recovery and 5% bioavailability was achieved after a single 50 mg/kg suspension dose. Similar values were obtained following a 250 mg/kg dose. Monkeys displayed a CI-976 elimination half-life of 0.6 hr after a 2 mg/kg intravenous dose. No unchanged drug was excreted in urine. In summary, CI-976 exhibits moderate absorption and bioavailability in the rat, but lower absorption and bioavailability in monkey. A special difference in CI-976 elimination half-life was observed consistent with in vitro metabolism results.


Subject(s)
Anilides/pharmacokinetics , Sterol O-Acyltransferase/antagonists & inhibitors , Administration, Oral , Anilides/blood , Anilides/urine , Animals , Biliary Tract/metabolism , Biological Availability , Carbon Radioisotopes , Feces/chemistry , Injections, Intravenous , Macaca fascicularis , Male , Rats , Rats, Wistar , Tissue Distribution
19.
Drug Metab Dispos ; 21(5): 874-82, 1993.
Article in English | MEDLINE | ID: mdl-7902251

ABSTRACT

Tacrine's [1,2,3,4-tetrahydro-9-acridinamine monohydrochloride monohydrate, (THA)] metabolic fate was examined using human and rat liver microsomal preparations. Following 1-hr incubations with human microsomes, [14C]THA (0.4 microM) was extensively metabolized to 1-hydroxyTHA with trace amounts of 2-, 4-, and 7-hydroxyTHA also produced. Poor recovery of radioactivity in the postreaction incubates suggested association of THA-derived radioactivity with precipitated microsomal protein. After exhaustive extraction, 0.034, 0.145, 0.126, and 0.012 nmol eq bound/mg protein/60 min of THA-derived radioactivity was bound to human liver preparations H109, H111, H116, and H118, respectively. Preparations H109 and H118 were lower in P4501A2 content and catalytic activity as compared with preparations H111 and H116. Incubations of equimolar [14C]1-hydroxyTHA with human liver microsomes also resulted in binding to protein, although to a lesser extent than observed with THA. [14C]THA (0.4 microM) was incubated for 1 hr with rat liver microsomes (1 microM P-450) prepared from noninduced (N), phenobarbital (PB), isoniazid (I), and 3-methylcholanthrene (3-MC)-pretreated animals. In all incubations, 1-hydroxyTHA was the major biotransformation product detected. After exhaustive extraction, 0.048, 0.054, 0.049, and 0.153 nmol eq/mg protein/60 min of THA-derived radioactivity was bound to microsomal protein from N, PB, I, and 3-MC pretreated rats. Increased binding with 3-MC induced rat liver preparations suggests the involvement of the P-450 1A subfamily in THA bioactivation. Glutathione (5 mM) coincubation inhibited the irreversible binding of THA-derived radioactivity in both human and 3-MC-induced rat liver preparations, whereas human epoxide hydrase (100 micrograms/incubate) had a relative minor effect. A mechanism is proposed involving a putative quinone methide(s) intermediate in the bioactivation and irreversible binding of THA. A species difference in THA-derived irreversible binding exists between human and noninduced rat liver microsomes, suggesting that the rat is a poor model for studying the underlying mechanism(s) of THA-induced elevations in liver marker enzymes found in clinical investigations.


Subject(s)
Microsomes, Liver/metabolism , Tacrine/metabolism , Tacrine/pharmacokinetics , Animals , Biotransformation , Carbon Radioisotopes , Cognition/drug effects , Epoxide Hydrolases/pharmacology , Glutathione/pharmacology , Humans , Male , Rats , Rats, Inbred Strains , Species Specificity , Tacrine/analogs & derivatives
20.
Biochem Pharmacol ; 46(1): 13-20, 1993 Jul 06.
Article in English | MEDLINE | ID: mdl-8347124

ABSTRACT

Tacrine (1,2,3,4-tetrahydro-9-aminoacridine hydrochloride; THA) is known to undergo extensive oxidative metabolism to a variety of mono- and dihydroxylated metabolites in animals and humans. The potential for tacrine to undergo metabolism to stable, protein-reactive and cytotoxic metabolites has been investigated in incubations with human and rat liver microsomes. Using lymphocytes as sensitive markers to quantify cytotoxicity, THA (50 microM) underwent NADPH-dependent bioactivation to a cytotoxic metabolite(s). NADPH-dependent cytotoxicity in the presence of rat and human microsomes was 9.8 +/- 3.1% (P < 0.05 cf. -NADPH control) and 6.2 +/- 2.0% (P < 0.05 cf. -NADPH control), respectively. Stable and protein-reactive metabolites were also formed in microsomes from both species. These accounted for 28.2 +/- 12.7% and 1.22 +/- 0.79% of incubated radioactivity in human microsomes and 6.4 +/- 2.2% and 0.4 +/- 0.1% of incubated radioactivity in rat microsomes. In microsomes pooled from six human livers the NADPH-dependent cytotoxicity was 9.4 +/- 1.1%. Formation of stable and protein-reactive metabolites accounted for 29.2 +/- 2.3% and 1.2 +/- 1.0% of incubated radioactivity. Reduced glutathione (500 microM) completely blocked NADPH-dependent cytotoxicity and inhibited protein-reactive metabolite formation by 60% (P < 0.05). Ascorbic acid (500 microM) inhibited the generation of cytotoxic and protein-reactive metabolites by 75% (P < 0.05) and 35% (P < 0.05), respectively. Cyclohexene oxide was without effect. Human serum albumin was found to protect the lymphocytes against toxicity. In microsomes prepared from the livers of four donors known to have been smokers there were no significant differences in the generation of metabolites from THA compared with microsomes prepared from livers of non-smokers. Enoxacin, a specific inhibitor of cytochrome P450 1A2 significantly inhibited all routes of THA metabolism. We have therefore demonstrated that THA may be oxidatively metabolized to stable, protein-reactive and cytotoxic metabolites in human and rat liver microsomes. A number of inhibitors may affect these process, whilst inhibition by enoxacin indicates a role for cytochrome P450 1A2 in THA metabolism.


Subject(s)
Microsomes, Liver/metabolism , Tacrine/metabolism , Adult , Animals , Ascorbic Acid/pharmacology , Biotransformation/drug effects , Cell Death/drug effects , Enoxacin/pharmacology , Glutathione/pharmacology , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , NADP/metabolism , Rats , Tacrine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...