Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Arch Biochem Biophys ; 396(1): 16-24, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11716457

ABSTRACT

Cytochrome P450 CYP2D6 metabolizes a wide range of pharmaceutical compounds. A CYP2D6 fusion enzyme (CYP2D6F), containing an amino-terminal human CYP2D6 sequence and a carboxyterminal human NADPH-cytochrome P450 oxidoreductase (CPR) moiety, was constructed. High levels of expression were achieved in Escherichia coli (60-100 nmol/liter) and the enzyme was catalytically active with optimal activities achieved in the presence of the antioxidant, GSH. Turnover values for bufuralol 1'-hydroxylation, metoprolol alpha-hydroxylation, O-desmethylation, and dextromethorphan O-demethylation, using membranes expressing the fusion enzyme, were 5.6, 0.4, 0.72, and 6.19 min(-1), respectively. These values were similar to E. coli membranes which coexpressed human CYP2D6 and CPR (CYP2D6/R). The K(m) and k(cat) values for bufuralol metabolism were estimated to be 10.2 microM and 4.1 min(-1), respectively. The enzyme was purified using ion-exchange chromatography, affinity chromatography (2'-5' ADP-Sepharose), and gel filtration. Estimated turnover rates for bufuralol 1'-hydroxylation, metoprolol alpha-hydroxylation, O-desmethylation, and dextromethorphan O-demethylation were 1.2, 0.52, 0.79, and 0.76 min(-1), respectively. Bufuralol 1'-hydroxylase activity by purified CYP2D6F was enhanced by phospholipids and added CPR. The CYP2D6F enzyme was able to stimulate CYP3A4 testosterone 6beta-hydroxylase activity in a reconstitution system indicating that electron transfer may be largely intermolecular. The catalytically self-sufficient CYP2D6F enzyme will facilitate investigations of P450-CPR interactions and the development of new biocatalysts.


Subject(s)
Cytochrome P-450 CYP2D6/metabolism , NADPH-Ferrihemoprotein Reductase/metabolism , Recombinant Fusion Proteins/metabolism , Cytochrome P-450 CYP2D6/genetics , Dextromethorphan/metabolism , Escherichia coli/genetics , Ethanolamines/metabolism , Humans , Metoprolol/metabolism , NADPH-Ferrihemoprotein Reductase/genetics , Pharmaceutical Preparations/metabolism , Protein Engineering , Technology, Pharmaceutical
2.
Br J Clin Pharmacol ; 48(3): 424-32, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10510156

ABSTRACT

AIMS: To identify the human cytochrome P450 enzyme(s) involved in the in vitro metabolism of rosiglitazone, a potential oral antidiabetic agent for the treatment of type 2 diabetes-mellitus. Method The specific P450 enzymes involved in the metabolism of rosiglitazone were determined by a combination of three approaches; multiple regression analysis of the rates of metabolism of rosiglitazone in human liver microsomes against selective P450 substrates, the effect of selective chemical inhibitors on rosiglitazone metabolism and the capability of expressed P450 enzymes to mediate the major metabolic routes of rosiglitazone metabolism. Result The major products of metabolism following incubation of rosiglitazone with human liver microsomes were para-hydroxy and N-desmethyl rosiglitazone. The rate of formation varied over 38-fold in the 47 human livers investigated and correlated with paclitaxel 6alpha-hydroxylation (P<0.001). Formation of these metabolites was inhibited significantly (>50%) by 13-cis retinoic acid, a CYP2C8 inhibitor, but not by furafylline, quinidine or ketoconazole. In addition, both metabolites were produced by microsomes derived from a cell line transfected with human CYP2C8 cDNA. There was some evidence for CYP2C9 playing a minor role in the metabolism of rosiglitazone. Sulphaphenazole caused limited inhibition (<30%) of both pathways in human liver microsomes and microsomes from cells transfected with CYP2C9 cDNA were able to mediate the metabolism of rosiglitazone, in particular the N-demethylation pathway, albeit at a much slower rate than CYP2C8. Rosiglitazone caused moderate inhibition of paclitaxel 6alpha-hydroxylase activity (CYP2C8; IC50=18 microm ), weak inhibition of tolbutamide hydroxylase activity (CYP2C9; IC50=50 microm ), but caused no marked inhibition of the other cytochrome P450 activities investigated (CYP1A2, 2A6, 2C9, 2C19, 2D6, 2E1, 3A and 4A). Conclusion CYP2C8 is primarily responsible for the hydroxylation and N-demethylation of rosiglitazone in human liver; with minor contributions from CYP2C9.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/metabolism , Steroid 16-alpha-Hydroxylase , Thiazoles/metabolism , Thiazolidinediones , Cytochrome P-450 CYP1A2 , Cytochrome P-450 CYP2C8 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 Enzyme System/genetics , DNA, Complementary/genetics , Enzyme Inhibitors/pharmacology , Humans , Hypoglycemic Agents/metabolism , Kinetics , Microsomes, Liver/enzymology , Rosiglitazone , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Transfection , Tumor Cells, Cultured
3.
Xenobiotica ; 29(3): 311-25, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10219970

ABSTRACT

1. The disposition and metabolic fate of ropinirole, a novel compound indicated for the symptomatic treatment of Parkinson's disease, was studied in the mouse, rat, cynomolgus monkey and man, following oral and intravenous administration of ropinirole hydrochloride. 2. In all species, nearly all of the p.o. administered dose (94%) was rapidly absorbed from the gastrointestinal tract following administration of 14C-ropinirole hydrochloride. In rat and monkey, the compound distributed rapidly beyond total body water and was shown to cross the blood-brain barrier. Blood clearance of the compound was high, approximately equal to one-half the hepatic blood flow in the monkey and similar to the hepatic blood flow in rat. Terminal phase elimination half-lives for the compound were relatively short (0.5 and 1.3 h in rat and monkey respectively), although there was evidence of a second elimination phase in the monkey with an elimination half-life of approximately 5-11 h. Plasma concentrations of ropinirole after the intravenous dose were not determined in the mouse and were below the lower limit of quantification in man (0.08 ng/ml) at the doses used in the studies described in this paper. 3. In both animals and man, ropinirole was extensively metabolized. In the rat, the major metabolic pathway was via hydroxylation of the aromatic ring to form 7-hydroxy ropinirole. In mouse, monkey and man, the major pathway was via N-depropylation. The N-despropyl metabolite was metabolized further to form 7-hydroxy and carboxylic acid derivatives. Metabolites formed in all species were generally metabolized further by glucuronidation. 7-Hydroxy ropinirole is the only metabolite of ropinirole previously shown to possess significant dopamine agonist activity in vivo. In all species, the major route of excretion of ropinirole-related material after oral or intravenous administration of the compound was renal (60-90% of dose).


Subject(s)
Antiparkinson Agents/metabolism , Dopamine Agonists/metabolism , Indoles/metabolism , Absorption , Adult , Animals , Antiparkinson Agents/pharmacokinetics , Carbon Radioisotopes , Dopamine/metabolism , Dopamine Agonists/pharmacokinetics , Dose-Response Relationship, Drug , Half-Life , Humans , Indoles/pharmacokinetics , Macaca fascicularis , Male , Mice , Mice, Inbred Strains , Middle Aged , Rats , Rats, Wistar , Reference Values , Species Specificity , Tissue Distribution
4.
Drug Metab Dispos ; 25(7): 840-4, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9224778

ABSTRACT

The in vitro metabolism of ropinirole was investigated with the aim of identifying the cytochrome P450 enzymes responsible for its biotransformation. The pathways of metabolism after incubation of ropinirole with human liver microsomes were N-despropylation and hydroxylation. Enzyme kinetics demonstrated the involvement of at least two enzymes contributing to each pathway. A high affinity component with a K(M) of 5-87 microM and a low affinity component with a K(M) of approximately two orders of magnitude greater were evident. The high affinity component could be abolished by pre-incubation of the microsomes with furafylline. Additionally, incubation of ropinirole with microsomes derived from CYP1A2 transfected cells readily produced the N-despropyl and hydroxy metabolites. Some inhibition of ropinirole metabolism was also observed with ketoconazole, indicating a minor contribution by CYP3A. Multivariate correlation data were consistent with the involvement of the cytochrome P450 enzymes 1A2 and 3A in the metabolism of ropinirole. Thus, it could be concluded that the major P450 enzyme responsible for ropinirole metabolism at lower (clinically relevant) concentrations is CYP1A2 with a contribution from CYP3A, particularly at higher concentrations.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Dopamine Agonists/pharmacokinetics , Indoles/pharmacokinetics , Isoenzymes/metabolism , Biotransformation , Cytochrome P-450 Enzyme Inhibitors , Dealkylation , Enzyme Inhibitors/pharmacology , Humans , Hydroxylation , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Multivariate Analysis
5.
Drug Metab Dispos ; 25(6): 757-61, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9193879

ABSTRACT

Rat liver microsomal testosterone (250 microM) hydroxylation and immunoreactive CYP3A protein were compared after administration of the antiglucocorticoid RU 486 (50 mg.kg-1.day-1 for 4 days) and the hypocholesterolaemic drug SR-12813 (150 mg.kg-1.day-1 for 4 days). Markers of CYP3A-mediated enzyme activity (testosterone 15 beta-, 6 beta-, and 2 beta-hydroxylation) were increased after administration of both drugs. Testosterone 6 beta-hydroxylation was increased 5-fold by RU 486 and 9-fold by SR-12813. Administration of dexamethasone alone at 150 mg.kg-1.day-1 or in combination with RU 486 induced testosterone 6 beta-hydroxylation 15- to 20-fold. The lack of antagonistic effect of RU 486 on dexamethasone-mediated CYP3A induction strengthens support for the hypothesis that the "classical glucocorticoid receptor" does not play a part in this process. The induction of CYP3A enzymes by the bisphosphonate SR-12813 suggests the existence of a new class of compounds with CYP3A inducing properties.


Subject(s)
Anticholesteremic Agents/pharmacology , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/biosynthesis , Diphosphonates/pharmacology , Hormone Antagonists/pharmacology , Liver/drug effects , Mifepristone/pharmacology , Oxidoreductases, N-Demethylating/biosynthesis , Animals , Blotting, Western , Cytochrome P-450 CYP3A , Enzyme Induction , Female , Liver/enzymology , Rats , Rats, Sprague-Dawley , Testosterone/metabolism
7.
Xenobiotica ; 25(9): 917-27, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8553685

ABSTRACT

1. Caffeine N3-demethylation, the major pathway of caffeine metabolism in man, is mediated by P4501A2. The carbon of the methyl group lost during N3-demethylation is eliminated as carbon dioxide in vivo, or as formaldehyde and formic acid in vitro. 2. A simple and sensitive assay was developed to quantify the [14C]formaldehyde/[14C]formic acid produced following incubation of human microsomes with [3-14C-methyl]caffeine. This assay, using solid-phase extraction, enables quantitation of [14C]formaldehyde/[14C]formic acid with acceptable precision (within 5%) and accuracy (within 10%). 3. Typical Km and Vmax for the N3-demethylation of caffeine were determined by this assay to be 500 (range 220-1200) microM, and 250 (range 115-450) pmol.mg protein-1.min-1 respectively. 4. The N3-demethylation activity determined in microsomes from a range of human livers correlated significantly with other P4501A2 activities (p < 0.001) and was inhibited (> 95%) by furafylline. In addition, caffeine N3-demethylation was catalysed by microsomes from cell lines transfected with human P4501A2 cDNA. 5. This assay, for quantitation of [14C]formaldehyde/[14C]formic acid in human liver microsomes, is suitable for use in in vitro drug interaction studies as a probe for P4501A2 activity.


Subject(s)
Caffeine/analogs & derivatives , Cytochrome P-450 Enzyme System/analysis , Microsomes, Liver/enzymology , Oxidoreductases/analysis , Carbon Radioisotopes , Cytochrome P-450 CYP1A2 , Formaldehyde/analysis , Formates/analysis , Humans , Radioligand Assay
8.
J Pharmacol Exp Ther ; 274(2): 761-6, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7636740

ABSTRACT

The kinetics of oxidation of phenytoin and tolbutamide were determined in freshly isolated hepatocytes and hepatic microsomes from male Sprague-Dawley rats. Similar enzyme kinetic models are applicable to the data from both in vitro systems; a two-site model for phenytoin with a high affinity (Km = 1-5 microM, based on unbound drug concentration), low capacity site and a low affinity, high capacity site, and a one-site model for tolbutamide. Steady-state infusion studies were performed to characterize the Michaelis-Menten parameters for phenytoin disposition in vivo, these data could also be described by a two-site metabolism model (Km 1.3 microM, intrinsic clearance 62 ml/min for unbound drug for the high affinity site). Comparison of in vivo and in vitro parameters (after scaling the latter parameters for either hepatocyte yield or microsomal recovery) showed excellent prediction of in vivo clearance of unbound drug from hepatocyte data (55 ml/min) but underprediction from microsomal data (17 ml/min). In contrast to phenytoin, the in vivo clearance of tolbutamide (1.5 ml/min for unbound drug) was equally well predicted by both hepatocyte (2.4 ml/min) and microsomal (3.1 ml/min) studies. The difference between the utility of in vitro systems to predict the in vivo clearance of these two drugs, which show similar pharmacrokinetic properties (low clearance restricted to unbound drug concentration in blood), may be a consequence of the particular terminal metabolite formed in each in vitro system.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Liver/metabolism , Microsomes, Liver/metabolism , Phenytoin/pharmacokinetics , Tolbutamide/pharmacokinetics , Animals , Liver/cytology , Male , Metabolic Clearance Rate , Rats , Rats, Sprague-Dawley
9.
Drug Metab Dispos ; 23(3): 349-53, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7628300

ABSTRACT

The kinetics of caffeine metabolism has been investigated in freshly isolated hepatocytes, hepatic microsomes, and in vivo in male Sprague-Dawley rats. A simple Michaelis-Menten model provides an adequate description of each of the three sets of data. There is reasonable agreement between the KM values for the three systems (56-200 microM). Vmax values for hepatocytes and microsomes show good agreement when expressed in the same units using scaling factors for hepatic cellularity and microsomal protein yield [315 and 420 nmol/min/standard rat weight (SRW), respectively]. Both values slightly exceed the in vivo-determined Vmax (190 nmol/min/SRW). Taking the Vmax/KM ratio (intrinsic clearance) as the basis for scaling, the in vitro data from both the hepatocyte (2.6 ml/min/SRW) and microsomal (2.7 ml/min/SRW) studies provide a good prediction of the in vivo total body clearance (3.4 ml/min/SRW).


Subject(s)
Caffeine/pharmacokinetics , Liver/metabolism , Microsomes, Liver/metabolism , Animals , In Vitro Techniques , Liver/cytology , Male , Metabolic Clearance Rate , Rats , Rats, Sprague-Dawley
10.
Xenobiotica ; 25(3): 261-70, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7618352

ABSTRACT

1. The potential of ketoconazole and sulphaphenazole to inhibit specific P450 enzyme activities (1A2, 2A6, 2B6, 2C9/8, 2C19, 2D6, 2E1, 3A and 4A) was investigated using human liver microsomes. 2. Ketoconazole demonstrated an inhibitory effect on cyclosporine oxidase and testosterone 6 beta-hydroxylase activities, with mean IC50's of 0.19 and 0.22 microM respectively. Ketoconazole inhibition of the other P450 activities investigated was significantly less, as illustrated by IC50's of at least a magnitude higher. 3. Sulphaphenazole was shown to have an inhibitory effect on tolbutamide hydroxylase activity, with a mean IC50 of 0.8 microM in incubations containing 100 microM tolbutamide. Sulphaphenazole (at concentrations of up to 100 microM) did not exhibit any significant inhibition of the other enzyme activities investigated. 4. Ketoconazole and sulphaphenazole are the respective selective inhibitors of P4503A and 2C9. Ketoconazole at 1 microM and sulphaphenazole at 10 microM can be used to establish the involvement of P4503A and 2C9 respectively in oxidative reactions in human liver microsomes.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme Inhibitors , Ketoconazole/pharmacology , Microsomes, Liver/enzymology , Mixed Function Oxygenases/antagonists & inhibitors , Sulfaphenazole/pharmacology , Cytochrome P-450 CYP2E1 , Female , Humans , Male , Steroid Hydroxylases/antagonists & inhibitors , Tolbutamide/metabolism
11.
Drug Metab Dispos ; 23(2): 251-4, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7736920

ABSTRACT

Famciclovir is the diacetyl 6-deoxy derivative of the active antiviral penciclovir that is for use in the treatment of infections caused by the herpes family of viruses. The major pathway of conversion is via di-deacetylation to BRL 42359, followed by oxidation to penciclovir. On oral dosing of famciclovir to humans, only penciclovir and BRL 42359 can be detected consistently in the plasma; thus, attention was focused on the oxidation reaction. This 6-oxidation occurred rapidly in human liver cytosol, had no requirement for cofactors, and followed simple Michaelis-Menten kinetics with a KM of 115 microM +/- 23 (N = 3). Using inhibitors of xanthine oxidase (allopurinol) and aldehyde oxidase (menadione and isovanillin), the relative roles of these enzymes in this process were determined. At a concentration of BRL 42359 that reflected plasma concentrations observed in humans (4 microM), both menadione (IC50 7 microM) and isovanillin (IC50 15 microM) caused extensive inhibition of the 6-oxidation reaction. In contrast, allopurinol caused no significant inhibition, confirming earlier in vivo work. At higher substrate concentrations (50 and 200 microM), the results with these inhibitors were broadly similar. These results provide strong evidence that aldehyde oxidase and not xanthine oxidase is responsible for the 6-oxidation of BRL 42359 to penciclovir in human liver cytosol, and this is likely to reflect the in vivo situation.


Subject(s)
2-Aminopurine/analogs & derivatives , Acyclovir/analogs & derivatives , Aldehyde Oxidoreductases/physiology , Antiviral Agents/metabolism , Liver/enzymology , Prodrugs/metabolism , 2-Aminopurine/metabolism , Acyclovir/metabolism , Aldehyde Oxidase , Aldehyde Oxidoreductases/antagonists & inhibitors , Allopurinol/pharmacology , Benzaldehydes/pharmacology , Cytosol/enzymology , Famciclovir , Guanine , Humans , Kinetics , Liver/drug effects , Oxidation-Reduction , Vitamin K/pharmacology , Xanthine Oxidase/antagonists & inhibitors , Xanthine Oxidase/metabolism
12.
Br J Clin Pharmacol ; 38(6): 557-66, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7888294

ABSTRACT

1. The metabolism of granisetron was investigated in human liver microsomes to identify the specific forms of cytochrome P450 responsible. 2. 7-hydroxy and 9'-desmethyl granisetron were identified as the major products of metabolism following incubation of granisetron with human liver microsomes. At low, clinically relevant, concentrations of granisetron the 7-hydroxy metabolite predominated. Rates of granisetron 7-hydroxylation varied over 100-fold in the human livers investigated. 3. Enzyme kinetics demonstrated the involvement of at least two enzymes contributing to the 7-hydroxylation of granisetron, one of which was a high affinity component with a Km of 4 microM. A single, low affinity, enzyme was responsible for the 9'-desmethylation of granisetron. 4. Granisetron caused no inhibition of any of the cytochrome P450 activities investigated (CYP1A2, CYP2A6, CYP2B6, CYP2C9/8, CYP2C19, CYP2D6, CYP2E1 and CYP3A), at concentrations up to 250 microM. 5. Studies using chemical inhibitors selective for individual P450 enzymes indicated the involvement of cytochrome P450 3A (CYP3A), both pathways of granisetron metabolism being very sensitive to ketoconazole inhibition. Correlation data were consistent with the role of CYP3A3/4 in granisetron 9'-desmethylation but indicated that a different enzyme was involved in the 7-hydroxylation.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Granisetron/metabolism , Isoenzymes/metabolism , Cytochrome P-450 Enzyme Inhibitors , Granisetron/pharmacology , Humans , Hydroxylation , In Vitro Techniques , Indoles/metabolism , Indoles/pharmacology , Isoenzymes/antagonists & inhibitors , Kinetics , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Ondansetron/metabolism , Ondansetron/pharmacology , Serotonin Antagonists/metabolism , Serotonin Antagonists/pharmacology , Tropisetron
13.
Chem Res Toxicol ; 7(6): 836-42, 1994.
Article in English | MEDLINE | ID: mdl-7696540

ABSTRACT

In vitro techniques have been utilized to investigate the microsomal enzymes involved in the metabolism of lauric acid and to establish conditions in which it can be used as a model substrate for both cytochrome P450 4A and cytochrome P450 2E1 in human liver microsomes. Studies of enzyme kinetics of lauric acid omega-hydroxylation in human liver microsomes indicated the involvement of more than one enzyme in this pathway, a relatively low Km enzyme with a Km of 22 microM +/- 12 (n = 8) and a high Km enzyme with a Km an order of magnitude higher (550 microM +/- 310, n = 7). The apparent Vmax for this component correlated with the rate of cyclosporin metabolism and was highly sensitive to ketoconazole inhibition. These results indicated that this enzyme was a member of the 3A subfamily. The activity associated with the low Km enzyme (P450 4A) did not correlate with P450 1A2, 2A6, 2C9/8, 2C19, 2D6, 2E1, or 3A activities in a bank of human liver microsomes and was not appreciably inhibited by ketoconazole, furafylline, quinidine, sulfaphenazole, or diethyldithiocarbamate (DDC). Lauric acid omega-1 hydroxylation demonstrated simple Michaelis-Menten kinetics in each of the human liver microsomal samples examined, with a Km of 130 microM +/- 42 (n = 8). This activity was highly correlated with chlorzoxazone 6-hydroxylation in human liver microsomes (r = 0.98, n = 14, p < 0.001) and was inhibited by both DDC and chlorzoxazone. Additionally, rats treated with the P450 2E1 inducer isoniazid demonstrated a 3-fold increase in lauric acid omega-1 hydroxylation relative to the control group. Thus, the lauric acid hydroxylation assay, at a substrate concentration of 20 microM, appears to be an effective and specific P450 model substrate capable of determining simultaneously P450 4A and P450 2E1 related activities in hepatic microsomal samples.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Lauric Acids/metabolism , Microsomes, Liver/enzymology , Mixed Function Oxygenases/metabolism , Oxidoreductases, N-Demethylating/metabolism , Animals , Cytochrome P-450 CYP2E1 , Cytochrome P-450 CYP4A , Female , Humans , Hydroxylation/drug effects , Ketoconazole/pharmacology , Kinetics , Male , Rats , Rats, Sprague-Dawley , Substrate Specificity
14.
Xenobiotica ; 24(6): 517-26, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7975717

ABSTRACT

1. Furafylline inhibition of 1A2-related activity in human liver microsomal systems was characterized. This inhibition was time and NADPH dependent. The kinetic constants were measured in human liver microsomes; a Ki of inactivation of 3 microM with a maximum rate constant of 0.27 min-1 were determined. 2. This inactivation process was retarded by the presence of a 1A2 substrate and after complete inhibition was achieved, 1A2 activity could be restored by the addition of fresh microsomes to the incubation mixture. These results are consistent with furafylline being a suicide substrate for 1A2. 3. Preincubating microsomes for 10 min with 10 microM furafylline in the presence of NADPH, prior to the initiation of the reaction by the addition of substrate, caused marked inhibition of 1A2 activity. This protocol was tested for specificity against 10 human P450 activities. The activities associated with 1A1, 2A6, 2B6, 2C9(/8), 2C19, 2D6, 2E1, 3A4(/5) and A were not significantly inhibited. 4. Using these conditions furafylline can be diagnostic of 1A2 involvement in a P450-dependent oxidative reaction.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Oxidoreductases/antagonists & inhibitors , Theophylline/analogs & derivatives , Cytochrome P-450 CYP1A1 , Cytochrome P-450 CYP1A2 , Cytochrome P-450 Enzyme System/metabolism , Humans , In Vitro Techniques , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , NADP/metabolism , Oxidoreductases/metabolism , Spectrophotometry, Ultraviolet , Theophylline/pharmacokinetics , Theophylline/pharmacology
16.
Drug Metab Dispos ; 22(2): 189-93, 1994.
Article in English | MEDLINE | ID: mdl-8013273

ABSTRACT

BRL 55792, BRL 55791, and BRL 55039 are prodrugs of an active antiviral agent 9-(3-hydroxypropoxy) guanine (BRL 44385). The prodrugs were 6-deoxygenated analogs of BRL 44385, with ether groups substituted at the 9-position: BRL 55792 with an (isopropoxymethyloxy)propoxy group, BRL 55791 with a (methoxymethyloxy)propoxy group, and BRL 55039 with an ethoxypropoxy group. Conversion of the prodrugs to BRL 44385 had been demonstrated in vivo in the rat and involved 6-oxidation followed by dealkylation. Metabolism was studied in rat liver in vitro systems to find a model to evaluate BRL 44385 production. Rat hepatocytes performed both reaction steps and were used to assess which of the three prodrugs demonstrated greatest production of the active drug. BRL 55792 demonstrated greatest conversion in vitro, and this was in agreement with in vivo data. The production of BRL 44385 from BRL 55792 was also demonstrated in human hepatocyte incubations, providing evidence that these reactions can occur in humans, thereby increasing confidence that BRL 55792 would be suitable prodrug for human therapy. Further experiments were performed to investigate the enzymes involved in these conversions. The 6-oxidation step occurred in the cytosol. Use of allopurinol and menadione (xanthine and aldehyde oxidase inhibitors) indicated that these conversions were catalyzed exclusively by xanthine oxidase in the rat but mainly by aldehyde oxidase in humans. The dealkylation reaction was detected in hepatocytes but not in homogenates or subcellular fractions. Inhibition of this reaction by aminobenzotriazole and ketoconazole (P-450 inhibitors) indicated that it was mediated by cytochrome P-450.


Subject(s)
Antiviral Agents/pharmacokinetics , Guanine/analogs & derivatives , Guanine/pharmacokinetics , Prodrugs/pharmacokinetics , Aldehyde Oxidoreductases/antagonists & inhibitors , Aldehyde Oxidoreductases/metabolism , Animals , Antiviral Agents/administration & dosage , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Cytosol/metabolism , Guanine/administration & dosage , Humans , In Vitro Techniques , Liver/metabolism , Microsomes, Liver/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Xanthine Oxidase/antagonists & inhibitors , Xanthine Oxidase/metabolism
17.
Drug Metab Dispos ; 22(1): 124-8, 1994.
Article in English | MEDLINE | ID: mdl-8149871

ABSTRACT

BRL 55792, BRL 55791, and BRL 55039 are prodrugs of an active anti-viral agent 9-(3-hydroxypropoxy) guanine, (BRL 44385). The prodrugs were 6-deoxygenated analogues of BRL 44385 with ether groups substituted at the 9-position: BRL 55792 with an (isopropoxymethyloxy)propoxy group, BRL 55791 with a (methoxymethyloxy)propoxy group, and BRL 55039 with an ethoxypropoxy group. Conversion of the prodrugs to BRL 44385 had been demonstrated in vivo in rat and involved 6-oxidation followed by dealkylation. Metabolism was studied in rat liver in vitro systems to find a model to evaluate BRL 44385 production. Rat hepatocytes performed both reaction steps and were used to assess which of the three prodrugs demonstrated greatest production of the active drug. BRL 55792 demonstrated greatest conversion in vitro and this was in agreement with in vivo data. The production of BRL 44385 from BRL 55792 was also demonstrated in human hepatocyte incubations providing evidence that these reactions can occur in man thereby increasing confidence that BRL 55792 would be a suitable prodrug for human therapy. Further experiments were performed to investigate the enzymes involved in these conversions. The 6-oxidation step occurred in the cytosol. Use of allopurinol and menadione (xanthine and aldehyde oxidase inhibitors) indicated that these conversions were catalyzed exclusively by xanthine oxidase in the rat but mainly by aldehyde oxidase in man. The dealkylation reaction was detected in hepatocytes but not in homogenates or subcellular fractions. Inhibition of this reaction by aminobenzotriazole and ketoconazole (P-450 inhibitors) indicated that it was mediated by cytochrome P-450.


Subject(s)
Antiviral Agents/metabolism , Guanine/analogs & derivatives , Guanine/metabolism , Prodrugs/metabolism , Animals , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Humans , Liver/cytology , Liver/enzymology , Models, Biological , Rats , Rats, Sprague-Dawley , Xanthine Oxidase/metabolism
18.
Drug Metab Dispos ; 21(1): 18-23, 1993.
Article in English | MEDLINE | ID: mdl-8095215

ABSTRACT

Famciclovir is the diacetyl 6-deoxy analog of the active antiviral compound penciclovir with potential use in the treatment of infections caused by the herpes family of viruses. The major pathway of metabolism of famciclovir is deacetylation to BRL 42359 followed by oxidation to penciclovir. It is possible that famciclovir may be coadministered with cyclosporin A to combat viral infections induced by immunosuppression in organ transplant and bone marrow transplant patients. As a result, information is required on possible interactions between the cytochrome P-450 3A substrate cyclosporin A and famciclovir and its metabolites in humans. In order to probe cytochrome P-450 3A activity, testosterone 6 beta-hydroxylation in two human liver microsomal preparations was measured. Nicardipine and ketoconazole, two drugs with known inhibitory interactions with cyclosporin A, were used as positive controls. Profiles of 6 beta-hydroxytestosterone production showed no inhibition effected by famciclovir, penciclovir, or BRL 42359 when marked inhibition was observed in incubations containing nicardipine, nifedipine, or ketoconazole. Further incubations of [14C]BRL 42359 with human liver cytosol and microsomes indicated that BRL 42359 is oxidized to penciclovir in cytosol but not in microsomes and that this reaction was not dependent on the presence of NADPH. Because P-450 resides mainly in the microsomal fraction and is dependent on the presence of cofactors for catalytic activity, it seems that this oxidation is not catalyzed by cytochrome P-450.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
2-Aminopurine/analogs & derivatives , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme Inhibitors , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Prodrugs/pharmacology , Testosterone/metabolism , 2-Aminopurine/metabolism , 2-Aminopurine/pharmacology , Acyclovir/analogs & derivatives , Acyclovir/metabolism , Acyclovir/pharmacology , Antiviral Agents/pharmacology , Carbon Radioisotopes , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Cytosol/metabolism , Famciclovir , Guanine , Humans , Hydroxylation/drug effects , In Vitro Techniques , Ketoconazole/pharmacology , Kinetics , Nicardipine/pharmacology , Nifedipine/pharmacology , Oxidoreductases, N-Demethylating/drug effects , Oxidoreductases, N-Demethylating/metabolism , Prodrugs/metabolism , Proteins/metabolism , Troleandomycin/pharmacology
19.
Biochem Pharmacol ; 42 Suppl: S17-25, 1991 Dec 11.
Article in English | MEDLINE | ID: mdl-1768275

ABSTRACT

Primary cultures of adult rat hepatocytes would provide a suitable system for the study of hepatic drug metabolism/toxicity provided that the drug-metabolizing enzymes could be maintained at levels approaching those seen in vivo. It has been reported that culture of adult rat hepatocytes in the presence of 2% (v/v) dimethyl sulphoxide (DMSO) allowed partial maintenance of total cytochrome P450 content. However, the levels of the individual isozymes were not determined. Culture of rat hepatocytes in the presence of DMSO did maintain the total cytochrome P450 content at 65% of the fresh cell value after 7 days of culture. This was accompanied by high cytochrome P420 levels suggesting that the solvent was stimulating de novo synthesis rather than maintaining existing enzyme. In the presence of DMSO the level of ethoxyresorufin-O-deethylase (EROD) rose 4-fold in culture, whilst that of pentoxyresorufin-O-dealkylase fell rapidly indicating that the isozyme pattern was altered significantly. The increases in total cytochrome P450 content and EROD activity were prevented by cycloheximide confirming that de novo protein synthesis was occurring. Haem oxygenase activity was significantly reduced and aminolaevulinic acid synthetase was significantly increased in the presence of solvent, suggesting increased haem availability for incorporation into cytochrome P450. However increased haem availability is insufficient in explaining the isozyme specificity of cytochrome P450 induction. Hepatocytes cultured in the presence of 2% (v/v) DMSO were markedly more responsive to 1,2-benzanthracene, with EROD increasing approximately 40-fold.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Dimethyl Sulfoxide/pharmacology , Isoenzymes/biosynthesis , Liver/enzymology , Oxidoreductases/biosynthesis , Animals , Benz(a)Anthracenes/toxicity , Cell Survival/drug effects , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Culture Media , Cytochrome P-450 CYP1A1 , Drug Synergism , Enzyme Induction/drug effects , Female , Heme Oxygenase (Decyclizing)/biosynthesis , Liver/drug effects , Mixed Function Oxygenases/biosynthesis , Porphobilinogen Synthase/biosynthesis , Rats , Rats, Inbred Strains
20.
Biochem Pharmacol ; 42(6): 1241-5, 1991 Aug 22.
Article in English | MEDLINE | ID: mdl-1888333

ABSTRACT

Diazepam metabolism has been investigated in rat hepatocytes cultured for 3, 24, 48 and 72 hr under five different conditions. Although four of the treatments studied reduced markedly the spontaneous loss of cytochrome P450, they had different effects on the metabolism of diazepam (DZ) presumably by affecting the relative proportions of cytochrome P450 isozymes during the period of culture. Thus P450 medium or dimethyl sulphoxide-supplemented medium maintained the rate of disappearance of DZ from the culture medium and metabolite profile in 24 hr cultures at the initial levels found in 3 hr cultures, while culture at 30 degrees or in metyrapone-containing medium resulted in the production of oxazepam, a metabolite normally only produced by dog, monkey and human hepatocytes. These findings indicate that the well recognized phenotypic alteration of cytochrome P450-dependent mono-oxygenase activities that occurs when rat hepatocytes are cultured in different media can result in a range of metabolic options that are normally only available in other animal species.


Subject(s)
Culture Media , Cytochrome P-450 Enzyme System/metabolism , Diazepam/metabolism , Liver/metabolism , Animals , Cells, Cultured , Dimethyl Sulfoxide , Hydrocortisone/pharmacology , Liver/enzymology , Male , Metyrapone/pharmacology , Models, Biological , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...