Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biopharm Drug Dispos ; 32(6): 303-18, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21725985

ABSTRACT

The 'relative activity factor' (RAF) compares the activity per unit of microsomal protein in recombinantly expressed cytochrome P450 enzymes (rhCYP) and human liver without separating the potential sources of variation (i.e. abundance of enzyme per mg of protein or variation of activity per unit enzyme). The dimensionless 'inter-system extrapolation factor' (ISEF) dissects differences in activity from those in CYP abundance. Detailed protocols for the determination of this scalar, which is used in population in vitro-in vivo extrapolation (IVIVE), are currently lacking. The present study determined an ISEF for CYP2C9 and, for the first time, systematically evaluated the effects of probe substrate, cytochrome b5 and methods for assessing the intrinsic clearance (CL(int) ). Values of ISEF for S-warfarin, tolbutamide and diclofenac were 0.75 ± 0.18, 0.57 ± 0.07 and 0.37 ± 0.07, respectively, using CL(int) values derived from the kinetic values V(max) and K(m) of metabolite formation in rhCYP2C9 + reductase + b5 BD Supersomes™. The ISEF values obtained using rhCYP2C9 + reductase BD Supersomes™ were more variable, with values of 7.16 ± 1.25, 0.89 ± 0.52 and 0.50 ± 0.05 for S-warfarin, tolbutamide and diclofenac, respectively. Although the ISEF values obtained from rhCYP2C9 + reductase + b5 for the three probe substrates were statistically different (p < 0.001), the use of the mean value of 0.54 resulted in predicted oral clearance values for all three substrates within 1.4 fold of the observed literature values. For consistency in the relative activity across substrates, use of a b5 expressing recombinant system, with the intrinsic clearance calculated from full kinetic data is recommended for generation of the CYP2C9 ISEF. Furthermore, as ISEFs have been found to be sensitive to differences in accessory proteins, rhCYP system specific ISEFs are recommended.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Microsomes, Liver/enzymology , Recombinant Proteins/metabolism , Anti-Inflammatory Agents, Non-Steroidal/analysis , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anticoagulants/analysis , Anticoagulants/metabolism , Aryl Hydrocarbon Hydroxylases/analysis , Cytochrome P-450 CYP2C9 , Cytochrome P-450 Enzyme System/analysis , Cytochrome P-450 Enzyme System/metabolism , Cytochromes b5/analysis , Cytochromes b5/metabolism , Diclofenac/analysis , Diclofenac/metabolism , Enzyme Assays , Forecasting/methods , Humans , Hypoglycemic Agents/analysis , Hypoglycemic Agents/metabolism , Liver/metabolism , Microsomes, Liver/metabolism , Models, Biological , Tolbutamide/analysis , Tolbutamide/metabolism , Warfarin/analysis , Warfarin/metabolism
2.
Article in English | MEDLINE | ID: mdl-17085084

ABSTRACT

A rapid, sensitive and selective LC-MS method is described for the simultaneous determination of zuclomiphene and enclomiphene in plasma from patients undergoing treatment with clomiphene citrate for the induction of ovulation. Samples spiked with N-didesmethyltamoxifen, the internal standard, were extracted into methyl tertiary butyl ether. The compounds were separated on a Luna C(18) analytical column, and a mobile phase of methanol-water (70:30 v/v) containing 0.05% trifluoroacetic acid at a flow rate of 1ml/min. The limits of determination were 35pg/ml and 7pg/ml for zu- and enclomiphene, respectively. Within-day coefficients of variation ranged from 2.1% to 7.2%.


Subject(s)
Chromatography, High Pressure Liquid/methods , Clomiphene/pharmacology , Enclomiphene , Mass Spectrometry/methods , Ovulation/drug effects , Clomiphene/blood , Clomiphene/chemistry , Female , Humans , Molecular Structure , Reproducibility of Results
4.
Xenobiotica ; 33(3): 225-37, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12637241

ABSTRACT

1. Interpretation of novel drug exposure and toxicology data from the dog is tempered by our limited molecular and functional knowledge of dog cytochromes P450 (CYPs). The aim was to study the mRNA and protein expression of hepatic dog CYPs in relation to the metabolism of substrates of human CYP, particularly those of the CYP2C subfamily. 2. The rate of 7-hydroxylation of S-warfarin (CYP2C9 in humans) by dog liver microsomes (mean +/- SD from 12 (six male and six female) dogs = 10.8 +/- 1.9 fmol mg(-1) protein min(-1)) was 1.5-2 orders of magnitude lower than that in humans. 3. The rate of 4'-hydroxylation of S-mephenytoin, catalysed in humans by CYP2C19, was also low in dog liver (4.6 +/-1.5 pmol mg(-1) protein min(-1)) compared with human liver. In contrast, the rate of 4'-hydroxylation of the R-enantiomer of mephenytoin by dog liver was much higher. The kinetics of this reaction (range of K(m) or K(0.5) 15-22 micro M, V(max) 35-59 pmol mg(-1) protein min(-1), n = 4 livers) were consistent with the involvement of a single enzyme. 4. In contrast to our findings for S-mephenytoin, dog liver microsomes 5'-hydroxylated omeprazole (also catalysed by CYP2C19 in humans) at considerably higher rates (range of K(m) 42-64 micro M, V(max) 22-46 pmol mg(-1) protein min(-1), n = 4 livers). 5. For all the substrates except omeprazole, a sex difference in their metabolism was observed in the dog (dextromethorphan N-demethylation: female range = 0.7-0.9, male = 0.4-0.8 nmol mg(-1) protein min(-1) (p < 0.02); S-warfarin 7-hydroxylation: female = 9-15.5, male = 8-12 fmol mg(-1) protein min(-1) (p < 0.02); R-mephenytoin 4'-hydroxylation: female = 16-35, male = 11.5-19 pmol mg(-1) protein min(-1) (p < 0.01); omeprazole 5'-hydroxylation: female = 15-20, male 13-22 pmol mg(-1) protein min(-1) (p < 0.2)). 6. All dog livers expressed mRNA and CYP3A12, CYP2B11, CYP2C21 proteins, with no sex differences being found. Expression of CYP2C41 mRNA was undetectable in the livers of six of 11 dogs. 7. Correlation analysis suggested that CYP2B11 catalyses the N-demethylation of dextromethorphan (mediated in humans by CYP3A) and the 4'-hydroxylation of mephenytoin (mediated in humans by CYP2C19) in the dog, and that this enzyme and CYP3A12 contribute to S-warfarin 7-hydroxylation (mediated in humans by CYP2C9). 8. In conclusion, we have identified a distinct pattern of hepatic expression of the CYP2C41 gene in the Alderley Park beagle dog. Furthermore, marked differences in the metabolism of human CYP2C substrates were observed in this dog strain compared with humans with respect to rate of reaction, stereoselectivity and CYP enzyme selectivity.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/metabolism , Dextromethorphan/analogs & derivatives , Liver/enzymology , Mephenytoin/analogs & derivatives , Protein Biosynthesis , RNA, Messenger/biosynthesis , Warfarin/analogs & derivatives , Algorithms , Animals , Dextromethorphan/metabolism , Dogs , Female , Humans , Isoenzymes/biosynthesis , Kinetics , Male , Mephenytoin/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Omeprazole/analogs & derivatives , Omeprazole/metabolism , Warfarin/metabolism
5.
Biochem Pharmacol ; 53(2): 171-8, 1997 Jan 24.
Article in English | MEDLINE | ID: mdl-9037249

ABSTRACT

4-Hydroxylation is an important pathway of tamoxifen metabolism because the product of this reaction is intrinsically 100 times more potent as an oestrogen receptor antagonist than is the parent drug. Although tamoxifen 4-hydroxylation is catalysed by human cytochrome P450 (CYP), data conflict on the specific isoforms responsible. The aim of this study was to define unequivocally the role of individual CYPs in the 4-hydroxylation of tamoxifen by human liver microsomes. Microsomes from each of 10 human livers catalysed the reaction [range = 0.6-2.9 pmol/mg protein/min (1 microM substrate concentration) and 6-25 pmol/mg protein/min (18 microM)]. Three of the livers with the lowest tamoxifen 4-hydroxylation activity were from genetically poor metabolisers with respect to CYP2D6. Inhibition of activity by quinidine (1 microM), sulphaphenazole (20 microM) and ketoconazole (2 microM), selective inhibitors of CYPs 2D6, 2C9 and 3A4, respectively, was 0-80%, 0-80% and 12-57%. The proportion of activity inhibited by quinidine correlated positively with total microsomal tamoxifen 4-hydroxylation activity (rs = 0.89, P < 0.01), indicating a major involvement of CYP2D6 in this reaction. Recombinant human CYPs 2D6, 2C9 and 3A4 but not CYPs 1A1, 1A2, 2C19 and 2E1 displayed significant 4-hydroxylation activity. Similar inhibition and correlation experiments confirmed that tamoxifen N-demethylation is catalysed predominantly by CYP3A4. These findings indicate that the 4-hydroxylation of tamoxifen is catalysed almost exclusively by CYPs 2D6, 2C9 and 3A4 in human liver microsomes. However, the marked between-subject variation in the contribution of these isoforms underlines the need to study metabolic reactions in a sufficient number of livers that are characterised with respect to a range of cytochrome P450 activities.


Subject(s)
Antineoplastic Agents, Hormonal/metabolism , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 CYP2D6/physiology , Cytochrome P-450 Enzyme System/physiology , Microsomes, Liver/metabolism , Mixed Function Oxygenases/physiology , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/physiology , Tamoxifen/metabolism , Adult , Cytochrome P-450 CYP3A , Dealkylation , Female , Humans , Hydroxylation , Male , Middle Aged
6.
Br J Clin Pharmacol ; 34(3): 262-5, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1389951

ABSTRACT

Inhibition of human cytochrome P4502D6 (CYP2D6)-catalysed metabolism can lead to clinically significant alterations in pharmacokinetics. Since there is evidence that the selective serotonin reuptake inhibitor (SSRI) class of antidepressant drugs might inhibit CYP2D6, the effects of five SSRIs on human liver microsomal CYP2D6 activity were compared with each other and with three tricyclic antidepressant drugs. On a molar basis, paroxetine was the most potent of the SSRIs at inhibiting the CYP2D6-catalysed oxidation of sparteine (Ki = 0.15 microM), although fluoxetine (0.60 microM) and sertaline (0.70 microM) had Ki values in the same range. Fluvoxamine (8.2 microM) and citalopram (5.1 microM) also inhibited CYP2D6 activity. The major circulating metabolites of paroxetine in man produced negligible inhibition. In contrast, norfluoxetine the active metabolite of fluoxetine, was a potent CYP2D6 inhibitor (0.43 microM). CYP2D6 activity was also diminished by the tricyclic antidepressant drugs clomipramine (2.2 microM), desipramine (2.3 microM) and amitriptyline (4.0 microM). These findings suggest that compounds with SSRI activity are likely to interact with human CYP2D6 in vivo with the potential of causing drug interactions.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Microsomes, Liver/enzymology , Mixed Function Oxygenases/antagonists & inhibitors , Selective Serotonin Reuptake Inhibitors/pharmacology , 1-Naphthylamine/analogs & derivatives , 1-Naphthylamine/pharmacology , Antidepressive Agents, Tricyclic/pharmacokinetics , Antidepressive Agents, Tricyclic/pharmacology , Citalopram/pharmacology , Cytochrome P-450 CYP2D6 , Drug Interactions , Fluoxetine/pharmacology , Fluvoxamine/pharmacology , Humans , Microsomes, Liver/drug effects , Paroxetine/pharmacology , Sertraline , Sparteine/metabolism
7.
J Pharm Pharmacol ; 43(10): 715-20, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1682447

ABSTRACT

The site of metabolic inversion of R(-)-ibuprofen to the pharmacologically active S(+)-enantiomer has been investigated using an array of in-situ rat perfused organ preparations allowing vascular perfusion (55-60 min) of the separate or combined intestine and liver. After addition of R(-)-ibuprofen (20 mg kg-1 body weight) to the closed (static) lumen of isolated 25 cm lengths of duodenum, jejunum or ileum, and single-pass vascular perfusion, both isomers were measured in the lumen and in vascular perfusate plasma (mean plasma AUC values (+/- s.d., micrograms mL-1 min, n = 5) R(-)-ibuprofen: 1669 +/- 115 (duodenum), 1687 +/- 203 (jejunum), 2061 +/- 188 (ileum); S(+)-ibuprofen: 23 +/- 6 (duodenum), 14 +/- 5 (jejunum), 26 +/- 1 (ileum]. Addition of the same dose of S(+)-ibuprofen to the jejunum (n = 5) resulted in AUC values of 1864 +/- 238 for S(+)-ibuprofen and 6 +/- 3 for R(-)-ibuprofen. After addition of R(-)-ibuprofen (30 micrograms mL-1) to the recirculating vascular perfusate (100 mL) of the entire small intestine (n = 6) AUC values were 1647 +/- 34 for R(-)-ibuprofen and 13 +/- 3 for S(-)-ibuprofen. The same dose of R(-)-ibuprofen to combined intestine/liver (n = 6) and liver only preparations (n = 6) gave AUC values of 1011 +/- 25 and 1021 +/- 49 for R(-)-ibuprofen and 220 +/- 28 and 238 +/- 22 for S(+)-ibuprofen, respectively. In all experiments, except those involving perfusion of the combined intestine/liver and the liver, the concentrations of the isomer opposite to that administered could be accounted for solely by the level of enantiomeric impurity (1.3% for R(-)-ibuprofen and 0.6% for S(+)-ibuprofen). We conclude that inversion of R(-)-ibuprofen to the S(+) antipode occurs in the liver but does not occur on either mucosal or serosal sides of the small intestine of the rat.


Subject(s)
Ibuprofen/metabolism , Intestinal Mucosa/metabolism , Liver/metabolism , Animals , Male , Perfusion , Rats , Rats, Inbred Strains , Stereoisomerism
8.
Biochem Pharmacol ; 38(16): 2639-44, 1989 Aug 15.
Article in English | MEDLINE | ID: mdl-2764987

ABSTRACT

The imidazole ring is a common structural feature of some xenobiotics that inhibit cytochrome P-450-catalysed reactions. Histamine is a 4-substituted imidazole and a preliminary study has shown it to be an inhibitor of rat liver microsomal drug oxidation. This work has now been extended. Histamine appears to be a competitive inhibitor of the alpha-hydroxylation (HM) (Ki = 164 microM; IC50 at 20 microM = 308 microM) and O-demethylation (ODM) (Ki = 243 microns; IC50 at 20 microM = 400 microM) of metoprolol in rat liver microsomes. Of the metabolites of histamine only N-acetylhistamine showed comparable inhibitory potency to that of the parent compound. Histamine impaired the disappearance of lignocaine when incubated with rat liver microsomes. This was accompanied by a corresponding inhibition of 3-hydroxy-lignocaine appearance. Histamine produced a type II spectral interaction with rat liver microsomes (lambda max = 432 nm, lambda min = 408 nm; Ks = 0.11 mM). When histamine was incubated alone with rat liver microsomes no loss of substrate was observed. The oxidation of metoprolol by human liver microsomes was impaired by histamine (IC50 values for ODM appearance at 25 microM: liver HL1 greater than 10, HL3 = 3.8 and HL4 = 3.7 mM). In comparison, cimetidine had an IC50 value of 1.5 mM using microsomes from liver HL3. Addition of histamine impaired the elimination of metoprolol by the isolated perfused rat liver in a dose-dependent manner (P less than 0.001, one-way analysis of variance). These data demonstrate that histamine can enter hepatocytes, interact with cytochrome P-450 and inhibit some drug oxidation reactions. The physiological relevance of inhibition of drug metabolism by histamine remains to be determined.


Subject(s)
Histamine/physiology , Microsomes, Liver/metabolism , Mixed Function Oxygenases/antagonists & inhibitors , Animals , Humans , In Vitro Techniques , Lidocaine/metabolism , Male , Metoprolol/metabolism , Rats
9.
J Pharmacol Exp Ther ; 247(1): 242-7, 1988 Oct.
Article in English | MEDLINE | ID: mdl-3171974

ABSTRACT

The oxidation of the beta adrenoceptor antagonist metoprolol exhibits genetic polymorphism of the sparteine/debrisoquine (SP/DB) type. The alpha-hydroxylation of metoprolol is absent in poor metabolizers, whereas metoprolol O-demethylation is only partially impaired, suggesting that an enzyme or enzymes other than cytochrome P450-SP/DB contribute to the latter reaction. Using inhibition by the quinidine/quinine isomer pair as a marker for the activity of cytochrome P450-SP/DB, the role of this enzyme in the in vitro oxidation of the enantiomers of metoprolol by human liver microsomes was examined. Unlike alpha-hydroxylation, only a portion of metoprolol O-demethylation showed the stereoselective inhibition by quinidine and quinine characteristic of in vitro reactions catalyzed by cytochrome P450-SP/DB. Furthermore, the kinetics of metoprolol O-demethylation were biphasic, the two components of O-demethylase activity being distinguishable by their enantioselectivity and sensitivity to inhibition by quinidine. Microsomes from one liver formed no detectable alpha-hydroxymetoprolol, and O-demethylation by these microsomes corresponded to the low affinity site observed in eight other livers. The rate of metoprolol O-demethylation by the quinidine-inhibitable high affinity component was directly proportional to the rate of alpha-hydroxylation. These findings support the hypothesis that cytochrome P450-SP/DB catalyzes the formation of alpha-hydroxymetoprolol, but is only partially responsible for metoprolol O-demethylation. Such a mechanism could explain the previously reported inability to detect polymorphism in the O-demethylation pathway in vivo.


Subject(s)
Cytochrome P-450 Enzyme System/physiology , Debrisoquin/metabolism , Isoquinolines/metabolism , Metoprolol/metabolism , Microsomes, Liver/metabolism , Quinidine/pharmacology , Sparteine/metabolism , Dealkylation , Humans , Hydroxylation , In Vitro Techniques , Kinetics , Quinine/pharmacology
10.
Biochem Pharmacol ; 35(16): 2757-61, 1986 Aug 15.
Article in English | MEDLINE | ID: mdl-2943287

ABSTRACT

The oxidative metabolism of metoprolol has been shown to display genetic polymorphism of the debrisoquine-type. The use of in vitro inhibition studies has been proposed as a means of defining whether one or more forms of cytochrome P-450 are involved in the monogenically-controlled metabolism of two substrates. We have, therefore, tested the ability of debrisoquine and other substrates to inhibit the oxidation of metoprolol by rat liver microsomes. Debrisoquine and guanoxan were potent competitive inhibitors of the alpha-hydroxylation and O-desmethylation of metoprolol as well as its metabolism by all routes (measured by substrate disappearance). Cimetidine and ranitidine, drugs which are known to impair the clearance of metoprolol in man, showed an inhibitory action comparable to that of debrisoquine in rat liver microsomes. Antipyrine, a compound whose metabolism is not impaired in poor metabolisers of debrisoquine, was found to be only a weak inhibitor of the metabolism of metoprolol. These findings suggest that the oxidation of metoprolol is linked closely to that of debrisoquine, cimetidine and ranitidine but not to that of antipyrine in the rat.


Subject(s)
Debrisoquin/pharmacology , Isoquinolines/pharmacology , Metoprolol/metabolism , Microsomes, Liver/metabolism , Animals , Antipyrine/pharmacology , Cimetidine/pharmacology , Guanidines/pharmacology , Ranitidine/pharmacology , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...