Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Br J Pharmacol ; 169(1): 51-68, 2013 May.
Article in English | MEDLINE | ID: mdl-23425116

ABSTRACT

BACKGROUND AND PURPOSE: AMG 181 is a human anti-α4 ß7 antibody currently in phase 1 and 2 trials in subjects with inflammatory bowel diseases. AMG 181 specifically targets the α4 ß7 integrin heterodimer, blocking its interaction with mucosal addressin cell adhesion molecule-1 (MAdCAM-1), the principal ligand that mediates α4 ß7 T cell gut-homing. EXPERIMENTAL APPROACH: We studied the in vitro pharmacology of AMG 181, and the pharmacokinetics and pharmacodynamics of AMG 181 after single or weekly i.v. or s.c. administration in cynomolgus monkeys for up to 13 weeks. KEY RESULTS: AMG 181 bound to α4 ß7 , but not α4 ß1 or αE ß7 , and potently inhibited α4 ß7 binding to MAdCAM-1 (but not vascular cell adhesion molecule-1) and thus inhibited T cell adhesion. Following single i.v. administration, AMG 181 Cmax was dose proportional from 0.01 to 80 mg·kg(-1) , while AUC increased more than dose proportionally. Following s.c. administration, dose-proportional exposure was observed with single dose ranging from 5 to 80 mg·kg(-1) and after 13 weekly doses at levels between 20 and 80 mg·kg(-1) . AMG 181 accumulated two- to threefold after 13 weekly 80 mg·kg(-1) i.v. or s.c. doses. AMG 181 had an s.c. bioavailability of 80%. The linear elimination half-life was 12 days, with a volume of distribution close to the intravascular plasma space. The mean trend for the magnitude and duration of AMG 181 exposure, immunogenicity, α4 ß7 receptor occupancy and elevation in gut-homing CD4+ central memory T cell count displayed apparent correlations. CONCLUSIONS AND IMPLICATIONS: AMG 181 has in vitro pharmacology, and pharmacokinetic/pharmacodynamic and safety characteristics in cynomolgus monkeys that are suitable for further investigation in humans.


Subject(s)
Antibodies, Monoclonal/administration & dosage , CD4-Positive T-Lymphocytes/metabolism , Immunoglobulins/metabolism , Integrins/metabolism , Mucoproteins/metabolism , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Biological Availability , CD4-Positive T-Lymphocytes/immunology , Cell Adhesion/drug effects , Cell Adhesion Molecules , Cell Line , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Injections, Intravenous , Injections, Subcutaneous , Macaca fascicularis , Male , Tissue Distribution
2.
Xenobiotica ; 33(10): 999-1011, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14555337

ABSTRACT

1. The antitumour agent bropirimine undergoes significant Phase II conjugation in vivo. Incubation of [14C]bropirimine with human liver microsomes resulted in the formation of a single product peak (M1) using high-performance liquid chromatography with radiochemical detection and was tentatively assigned as bropirimine glucuronide based on sensitivity to beta-glucuronidase and by obtaining the expected mass of 442/444 amu with liquid chromatography/mass spectrometry. Following metabolite isolation, the structure of M1 was established as bropirimine O-glucuronide by 1H-nuclear magnetic spectroscopy. 2. Studies aimed at identifying the human liver UDP-glucuronosyltransferase (UGT) enzyme(s) involved in the glucuronidation of bropirimine were carried out using recombinant human UGTs and it was determined that glucuronidation of bropirimine was catalysed by UGT1A1, UGT1A3 and UGT1A9. Bropirimine O-glucuronidation followed Michaelis-Menten kinetics and the Km and Vmax (mean +/- SD; n = 3) were 1217 +/- 205 microM and 667 +/- 188 pmol min(-1) mg(-1), respectively. 3. The activity of bropirimine O-glucuronidation by human liver microsomes was inhibited by bilirubin (40%) and with mefenamic acid (80%). Although buprenorphine extensively inhibited the activity of bropirimine O-glucuronidation by UGT1A3, the inhibition profile did not parallel that observed in HLMs. 4. The results demonstrate that UGT1A9 and to a lesser extent UGT1A1 are responsible for the majority of bropirimine O-glucuronidation in man.


Subject(s)
Cytosine/analogs & derivatives , Cytosine/metabolism , Glucuronidase/metabolism , Microsomes, Liver/metabolism , Antineoplastic Agents/pharmacology , Chromatography, High Pressure Liquid , Chromatography, Liquid , Enzyme Inhibitors/pharmacology , Glucuronosyltransferase/antagonists & inhibitors , Glucuronosyltransferase/metabolism , Humans , Kidney/metabolism , Kinetics , Magnetic Resonance Spectroscopy , Mass Spectrometry , Microsomes/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Models, Chemical , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Time Factors , UDP-Glucuronosyltransferase 1A9
3.
J Pharmacol Toxicol Methods ; 45(1): 79-84, 2001.
Article in English | MEDLINE | ID: mdl-11489668

ABSTRACT

The cytochromes P-450 recognize and metabolize a broad range of structurally diverse therapeutic agents. As a consequence, many clinically relevant drug--drug interactions (DDI) are associated with inhibition and/or induction of a specific P-450 enzymes (in particular human cytochrome P-450 3A4, CYP3A4). In addition to inhibition and induction, CYP-mediated drug metabolism may be enhanced upon coincubation with certain compounds. Moreover, some of these enzyme-based interactions appear to be substrate specific. In this presentation, several issues associated with the generation of accurate DDI information will be discussed.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Mixed Function Oxygenases/antagonists & inhibitors , Algorithms , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Enzyme Induction , Humans
4.
Arch Biochem Biophys ; 391(1): 49-55, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11414684

ABSTRACT

The inhibition of CYP3A4-mediated oxidation of triazolam and testosterone was assessed in the presence of a selection of known CYP3A4 substrates and inhibitors. Under experimental conditions where the Michaelis-Menten model predicts substrate-independent inhibition ([S] = K(m)), results yielded substrate-dependent inhibition. Moreover, when the same experimental design was extended to a group of structurally similar flavonoids it was observed that flavanone, flavone, 3-hydroxyflavone, and 6-hydroxyflavone (10 microM) activated triazolam metabolism, but inhibited testosterone hydroxylation. In additional studies, residual CYP3A4 activity toward testosterone and triazolam hydroxylation was measured after pretreatment with the CYP3A4 mechanism based inhibitor, midazolam. After midazolam preincubation, CYP3A4 6 beta-hydroxylase activity was reduced by 47% while, in contrast, triazolam hydroxylation was reduced by 75%. These results provide physical evidence, which supports the hypothesis that the active site of CYP3A4 contains spatially distinct substrate-binding domains within the enzyme active site.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Mixed Function Oxygenases/metabolism , Testosterone/metabolism , Triazolam/metabolism , Binding Sites , Binding, Competitive , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Midazolam/pharmacology , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/chemistry , Protein Structure, Tertiary , Substrate Specificity
5.
Drug Metab Dispos ; 29(1): 41-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11124228

ABSTRACT

Delavirdine, a non-nucleoside inhibitor of HIV-1 reverse transcriptase, is metabolized primarily through desalkylation catalyzed by CYP3A4 and CYP2D6 and by pyridine hydroxylation catalyzed by CYP3A4. It is also an irreversible inhibitor of CYP3A4. The interaction of delavirdine with CYP2C9 was examined with pooled human liver microsomes using diclofenac 4'-hydroxylation as a reporter of CYP2C9 catalytic activity. As delavirdine concentration was increased from 0 to 100 microM, the K(M) for diclofenac metabolism rose from 4.5+/-0.5 to 21+/-6 microM, and V(max) declined from 4.2+/-0.1 to 0.54+/-0.08 nmol/min/mg of protein, characteristic of mixed-type inhibition. Nonlinear regression analysis revealed an apparent K(i) of 2.6+/-0.4 microM. There was no evidence for bioactivation as prerequisite to inhibition of CYP2C9. Desalkyl delavirdine, the major circulating metabolite of delavirdine, had no apparent effect on microsomal CYP2C9 activity at concentrations up to 20 microM. Several analogs of delavirdine showed similar inhibition of CYP2C9. Delavirdine significantly inhibited cDNA-expressed CYP2C19-catalyzed (S)-mephenytoin 4'-hydroxylation in a noncompetitive manner, with an apparent K(i) of 24+/-3 microM. Delavirdine at concentrations up to 100 microM did not inhibit the activity of CYP1A2 or -2E1. Delavirdine competitively inhibited recombinant CYP2D6 activity with a K(i) of 12.8+/-1.8 microM, similar to the observed K(M) for delavirdine desalkylation. These results, along with previously reported experiments, indicate that delavirdine can partially inhibit CYP2C9, -2C19, -2D6, and -3A4, although the degree of inhibition in vivo would be subject to a variety of additional factors.


Subject(s)
Anti-HIV Agents/pharmacology , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 CYP2D6 Inhibitors , Cytochrome P-450 Enzyme Inhibitors , Delavirdine/pharmacology , Microsomes, Liver/drug effects , Mixed Function Oxygenases/antagonists & inhibitors , Reverse Transcriptase Inhibitors/pharmacology , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/antagonists & inhibitors , Cytochrome P-450 CYP1A2 , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2C9 , Diclofenac/pharmacokinetics , Humans , Hydroxylation , Microsomes, Liver/enzymology , Recombinant Proteins/antagonists & inhibitors
6.
Drug Metab Dispos ; 29(1): 70-5, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11124232

ABSTRACT

In human liver microsomes, triazolam is principally metabolized by CYP3A4 to form two metabolites, 1'-hydroxytriazolam (1'OHTz) and 4-hydroxytriazolam (4OHTz). The velocity of 1'OHTz formation was found to decrease at higher triazolam concentrations (>200 microM), indicative of "substrate inhibition". Coincubation of [(14)C]triazolam with authentic metabolite standards of either 1'OHTz or 4OHTz up to 30 microM did not significantly inhibit the rate of [(14)C]1'OHTz formation. The effects of secondary compounds on triazolam oxidation were shown to be product-specific, producing either activation or inhibition depending on the triazolam metabolite monitored. When human liver microsomes were supplemented with exogenous human cytochrome b(5), it was observed that substrate inhibition was attenuated and the resulting increase in 1'OHTz formation, relative to control (nonsupplemented) incubations, corresponded to a decrease in the ratio of 4OHTz to 1'OHTz. In contrast, when cofactor (e.g., 100 microM NADPH) was rate limiting, the metabolite ratio (4OHTz/1'OHTz) was markedly increased over the entire substrate concentration range (0.5-1000 microM). To explain these kinetic observations, a two-site binding model is proposed in which triazolam is hypothesized to bind within the CYP3A4 active site in spatially distinct orientations, which may lead to the formation of either the 1'-hydroxytriazolam or 4-hydroxytriazolam. Differential inhibition/activation is consistent with this two-site model and substrate inhibition is hypothesized to result from competition between the two sites for reactive oxygen.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Heme/metabolism , Mixed Function Oxygenases/metabolism , Oxygen/metabolism , Binding Sites , Binding, Competitive , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Humans , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Mixed Function Oxygenases/antagonists & inhibitors , Triazolam/pharmacokinetics
8.
Xenobiotica ; 30(9): 905-14, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11055268

ABSTRACT

1. Roquinimex, a novel immunomodulator, is metabolized in liver microsomes from mouse and rat via cytochrome P450s to four hydroxylated and two demethylated metabolites (R1-6). The study investigated which cytochrome P450 enzyme(s) is responsible for the metabolism of roquinimex in man. 2. Enzyme kinetic analysis demonstrated an apparent Km = 1.28-7.00 mM and Vmax = 50-159 pmol x mg(-1) microsomal protein x min(-1) for the primary metabolites in human liver microsomes. The sum of Cl(int) for the primary pathways was 0.167 microl x mg(-1) microsomal protein x min(-1). 3. A correlation between the formation rate of R1-6 and 6beta-hydroxylation of testosterone was obtained within a panel of liver microsomes from 11 individuals (r2 = 0.72-0.97). Furthermore, significant inhibition (>90%) of roquinimex primary metabolism was demonstrated by ketoconazole and troleandomycin, specific inhibitors of CYP3A4 as well as with anti-CYP3A4 antibodies. Moreover, a similar metabolite pattern was produced from roquinimex by incubation with cDNA-expressed CYP3A4 as by human liver microsomes. 4. In conclusion, these data indicate a major role for CYP3A4 in the formation of roquinimex primary metabolites in human liver microsomes.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Hydroxyquinolines/metabolism , Microsomes, Liver/enzymology , Mixed Function Oxygenases/metabolism , Antibodies/pharmacology , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , DNA, Complementary/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression , Humans , Hydroxylation , Ketoconazole/pharmacology , Kinetics , Male , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/genetics , Testosterone/metabolism , Troleandomycin/pharmacology
9.
Drug Metab Dispos ; 28(10): 1198-201, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10997940

ABSTRACT

Phenyldiazene reacted with lymphoblast-expressed CYP3A4 to give a stable phenyl-iron complex that could be induced to rearrange in situ producing approximately equal amounts of four N-phenyl-protoporphyrin IX isomers (N(B):N(A):N(C):N(D), 01:01:02:02). In the presence of 10 mM MgCl(2), the formation profile of the protoporphyrin isomers was markedly altered compared with control, favoring the N(A) isomer (N(B):N(A):N(C):N(D), 01:34:01:02). In addition, an investigation of MgCl(2) effects on CYP3A4-mediated metabolism of triazolam revealed that 10 mM MgCl(2) increased the apparent K(m) of triazolam 4-hydroxylation from 83 to 173 microM and reduced the V(max) for the reaction from 3.4 to 2.4 min(-1). Moreover, when the reaction kinetics of the oxidation of pyrene by CYP3A4 was examined in the absence of MgCl(2), it was found that the substrate-velocity curve was best approximated by a sigmoidal velocity curve (Hill coefficient 1.7 +/- 0.1). However, when the reaction was conducted in the presence of 10 mM MgCl(2), the resulting pyrene kinetics was not sigmoidal but rather biphasic (Hill coefficient 0.80 +/- 0.07). Based on the current results, it appears that CYP3A4 is conformationally sensitive to its in vitro environment and parameters, such as the presence of a divalent magnesium, can have a measurable effect on active site topography and consequently catalytic activity.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , Magnesium/pharmacology , Mixed Function Oxygenases/chemistry , Binding Sites/drug effects , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Humans , Hydroxylation/drug effects , Imines/metabolism , Kinetics , Mixed Function Oxygenases/metabolism , Oxidation-Reduction/drug effects , Protein Conformation/drug effects , Protoporphyrins/metabolism , Pyrenes/metabolism , Spectrometry, Mass, Electrospray Ionization , Substrate Specificity , Triazolam/metabolism
10.
Drug Metab Dispos ; 28(9): 1014-7, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10950842

ABSTRACT

In vitro studies were conducted to identify the hepatic enzyme(s) responsible for the oxidative metabolism of linezolid. In human liver microsomes, linezolid was oxidized to a single metabolite, hydroxylinezolid (M1). Formation of M1 was determined to be dependent upon microsomal protein and NADPH. Over a concentration range of 2 to 700 microM, the rate of M1 formation conformed to first-order (nonsaturable) kinetics. Application of conventional in vitro techniques were unable to identify the molecular origin of M1 based on the following experiments: a) inhibitor/substrates for various cytochrome P-450 (CYP) enzymes were unable to inhibit M1 formation; b) formation of M1 did not correlate (r(2) < 0.23) with any of the measured catalytic activities across a population of human livers (n = 14); c) M1 formation was not detectable in incubations using microsomes prepared from a baculovirus insect cell line expressing CYPs 1A1, 1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, 3A4, 3A5, and 4A11. In addition, results obtained from an in vitro P-450 inhibition screen revealed that linezolid was devoid of any inhibitory activity toward the following CYP enzymes (CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4). Additional in vitro studies excluded the possibility of flavin-containing monooxygenase and monoamine oxidase as potential enzymes responsible for metabolite formation. However, metabolite formation was found to be optimal under basic (pH 9.0) conditions, which suggests the potential involvement of either an uncharacterized P-450 enzyme or an alternative microsomal mediated oxidative pathway.


Subject(s)
Acetamides/pharmacokinetics , Anti-Infective Agents/pharmacokinetics , Microsomes, Liver/metabolism , Oxazoles/pharmacokinetics , Oxazolidinones , Animals , Carbon Radioisotopes , Cell Line , Chromatography, High Pressure Liquid , Cytochrome P-450 Enzyme System/metabolism , Humans , Linezolid , Mass Spectrometry , Microsomes/enzymology , Oxidation-Reduction
11.
Chem Res Toxicol ; 13(4): 262-70, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10775326

ABSTRACT

7-Ethynylcoumarin was synthesized as a potential mechanism-based inhibitor, and it was found to be an effective inactivator of 7-ethoxy-4-(trifluoromethyl)coumarin (7EFC) O-deethylation catalyzed by purified, reconstituted P450 2B1. In contrast, 7-ethynylcoumarin demonstrated minimal inactivation of P450 2A6-mediated 7-hydroxycoumarin formation. The inactivation of P450 2B1 demonstrated pseudo-first-order kinetics and was NADPH- and inhibitor-dependent. The maximal rate constant for the inactivation of 2B1 was 0.39 min(-)(1) at 30 degrees C, and thus, the time required to inactivate 50% of the P450 2B1 that was present (t(1/2)) was 1.8 min. The estimated concentration which led to half-maximal inactivation (K(I)) was 25 microM. No protection from inactivation was seen in the presence of nucleophiles (glutathione and sodium cyanide), an iron chelator (deferroxamine), or superoxide dismutase and catalase. Addition of the substrate (7EFC) protected P450 2B1 from inactivation, in a concentration-dependent manner. The partition ratio for P450 2B1 was 25; i.e., the number of metabolic events was 25-fold higher than the number of inactivating events. Incubations of 7-ethynylcoumarin with P450 2B1 for 10 min resulted in an 80% loss in enzymatic activity, while 90% of the ability to form a reduced-CO complex remained. This activity loss was not recovered following dialysis, indicative of irreversible inactivation. Covalent attachment of the entire inhibitor and oxygen to apo-P450 2B1, in a 1:1 ratio, was shown via electrospray ion trap mass spectrometry. This method also verified the absence of modification to the heme or the cytochrome P450 reductase. Taken together, the characterization of the inhibition seen with P450 2B1 and 7-ethynylcoumarin was consistent with all of the criteria required to distinguish a mechanism-based inactivator. In addition, electrospray ion trap mass spectrometry has the potential to be applied to protein adducts above and beyond those associated with the mechanism-based inactivation of cytochrome P450s.


Subject(s)
Apoenzymes/metabolism , Coumarins/pharmacology , Cytochrome P-450 CYP2B1/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Animals , Coumarins/metabolism , Cytochrome P-450 CYP2B1/metabolism , Male , Mass Spectrometry , Rats , Rats, Inbred F344
12.
Drug Metab Dispos ; 27(11): 1334-40, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10534319

ABSTRACT

In vitro studies were conducted to identify the hepatic cytochrome P-450 (CYP) enzymes responsible for the oxidative metabolism of the individual enantiomers of reboxetine. In human liver microsomes, each reboxetine enantiomer was metabolized to one primary metabolite, O-desethylreboxetine, and three minor metabolites, two arising via oxidation of the ethoxy aromatic ring and a third yet unidentified metabolite. Over a concentration range of 2 to 200 microM, the rate O-desethylreboxetine formation for either enantiomer conformed to monophasic Michaelis-Menten kinetics. Evidence for a principal role of CYP3A in the formation of O-desethylreboxetine for (S, S)-reboxetine and (R,R)-reboxetine was based on the results from the following studies: 1) inhibition of CYP3A activity by ketoconazole markedly decreased the formation of O-desethylreboxetine, whereas inhibitors selective for other CYP enzymes did not inhibit reboxetine metabolism, 2) formation of O-desethylreboxetine correlated (r(2) = 0.99; p <.001) with CYP3A-selective testosterone 6-beta-hydroxylase activity across a population of human livers (n = 14). Consistent with inhibition and correlation data, O-desethylreboxetine formation was only detectable in incubations using microsomes prepared from a Baculovirus-insect cell line expressing CYP3A4. Furthermore, the apparent K(M) for the O-desethylation of reboxetine in cDNA CYP3A4 microsomes was similar to the affinity constants determined in human liver microsomes. In addition, (S,S)-reboxetine and (R,R)-reboxetine were found to be competitive inhibitors of CYP2D6 and CYP3A4 (K(i) = 2.5 and 11 microM, respectively). Based on the results of the study, it is concluded that the metabolism of both reboxetine enantiomers in humans is principally mediated via CYP3A.


Subject(s)
Antidepressive Agents/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/metabolism , Morpholines/pharmacokinetics , Antidepressive Agents/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Humans , Morpholines/pharmacology , Reboxetine , Recombinant Proteins/metabolism , Stereoisomerism
13.
J Clin Pharmacol ; 39(3): 260-7, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10073325

ABSTRACT

This study assessed whether the previously reported difference in tirilazad clearance between pre- and postmenopausal women is reversed by hormone replacement and whether this observation can be explained by differences in CYP3A4 activity. Ten healthy women from each group were enrolled: premenopausal (ages 18-35), postmenopausal (ages 50-70), postmenopausal receiving estrogen, and postmenopausal women receiving estrogen and progestin. Volunteers received 0.0145 mg/kg midazolam and 3.0 mg/kg tirilazad mesylate intravenously on separate days. Plasma tirilazad and midazolam were measured by HPLC/dual mass spectrophotometry (MS/MS) assays. Tirilazad clearance was significantly higher in premenopausal women (0.51 +/- 0.09 L/hr/kg) than in postmenopausal groups (0.34 +/- 0.07, 0.32 +/- 0.06, and 0.36 +/- 0.08 L/hr/kg, respectively) (p = 0.0001). Midazolam clearance (0.64 +/- 0.12 L/hr/kg) was significantly higher in premenopausal women compared to postmenopausal groups (0.47 +/- 0.11, 0.49 +/- 0.11, and 0.53 +/- 0.19 L/hr/kg, respectively) (p = 0.037). Tirilazad clearance was weakly correlated with midazolam clearance (r2 = 0.129, p = 0.02). Tirilazad clearance is faster in premenopausal women than in postmenopausal women, but the effect of menopause on clearance is not reversed by hormone replacement. Tirilazad clearance in these women is weakly related to midazolam clearance, a marker of CYP3A activity.


Subject(s)
Antioxidants/pharmacokinetics , Aryl Hydrocarbon Hydroxylases , Estrogens/pharmacology , Hormone Replacement Therapy , Pregnatrienes/pharmacokinetics , Progesterone/pharmacology , Adolescent , Adult , Age Factors , Aged , Area Under Curve , Body Weight , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/physiology , Drug Therapy, Combination , Estrogens/therapeutic use , Female , Humans , Metabolic Clearance Rate , Midazolam/analogs & derivatives , Midazolam/blood , Midazolam/pharmacokinetics , Middle Aged , Oxidoreductases, N-Demethylating/physiology , Postmenopause/metabolism , Pregnatrienes/blood , Premenopause/metabolism , Progesterone/therapeutic use
14.
J Pharmacol Exp Ther ; 287(2): 583-90, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9808684

ABSTRACT

Tirilazad mesylate (FREEDOX), a potent inhibitor of membrane lipid peroxidation in vitro, is under clinical development for the treatment of subarachnoid hemorrhage. In humans, tirilazad is cleared almost exclusively via hepatic elimination with a medium-to-high extraction ratio. In human liver microsomal preparations, tirilazad is biotransformed to multiple oxidative products and one reduced, pharmacologically active metabolite, U-89678. Characterization of the reduced metabolite by mass spectrometry and cochromatography with an authentic standard demonstrated that U-89678 was formed via stereoselective reduction of the Delta4 bond in the steroid A-ring. Kinetic analysis of tirilazad reduction in human liver microsomes revealed that kinetically distinct type 1 and type 2 5alpha-reductase enzymes were responsible for U-89678 formation; the apparent KM values for type 2 and type 1 were approximately 15 and approximately 0.5 microM, respectively. Based on pH dependence and finasteride inhibition studies, it was inferred that 5alpha-reductase type 1 was the high affinity/low capacity microsomal reductase that contributed to tirilazad clearance in vivo. In addition, a role for CYP3A4 in the metabolism of U-89678 was established using cDNA expressed CYP3A4 and correlation studies comparing U-89678 consumption with cytochrome P450 activities across a population of human liver microsomes. Collectively, these data suggest that formation of U-89678, a circulating pharmacologically active metabolite, contributes to the total metabolic elimination of tirilazad in humans and that clearance of U-89678 is mediated primarily via CYP3A4 metabolism. Therefore, concurrent administration of therapeutic agents that modulate 5alpha-reductase type 1 or CYP3A activity are anticipated to affect the pharmacokinetics of PNU-89678.


Subject(s)
Isoenzymes/metabolism , Microsomes, Liver/enzymology , Oxidoreductases/metabolism , Pregnatrienes/pharmacokinetics , Biotransformation , Cholestenone 5 alpha-Reductase , Chromatography, High Pressure Liquid , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacokinetics , Humans , Hydrogen-Ion Concentration , Mass Spectrometry , Neuroprotective Agents/pharmacokinetics , Oxidoreductases/antagonists & inhibitors
15.
J Pharmacol Exp Ther ; 287(2): 591-7, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9808685

ABSTRACT

The effect of oral finasteride, an inhibitor of 5alpha-reductase, on the clearance of tirilazad, a membrane lipid peroxidation inhibitor, was assessed in eight healthy men who received: 1) 10 mg/kg tirilazad mesylate solution orally on the 7th day of a 10-day regimen of 5 mg finasteride once daily, 2) 10 mg/kg tirilazad mesylate orally, 3) 2 mg/kg tirilazad mesylate i.v. on the 7th day of a 10-day regimen of 5 mg finasteride once daily and 4) 2 mg/kg tirilazad mesylate i.v., in a four-way cross-over design. Plasma concentrations of tirilazad and its active reduced metabolites (U-89678 and U-87999) were measured by liquid chromatography with tandem mass spectrometry (LC-MS-MS). Finasteride increased mean tirilazad areas under the curve by 21 and 29% for i.v. and p.o. tirilazad, respectively. Mean U-89678 areas under the curve were decreased 92 and 75% by finasteride administration with i.v. and p.o. tirilazad, respectively, and decreases of 94 and 85% in mean U-87999 area under the curve values were observed. These differences were statistically significant. These results indicate that finasteride inhibits the metabolism of tirilazad to U-89678. However, this inhibition has only a moderate effect on the overall clearance of tirilazad. These results thus confirm earlier in vitro work that showed that tirilazad is predominantly metabolized by CYP3A4. Although the major circulating metabolites of tirilazad are formed via reduction, this represents a minor route of tirilazad elimination in man.


Subject(s)
Enzyme Inhibitors/pharmacology , Finasteride/pharmacology , Oxidoreductases/antagonists & inhibitors , Pregnatrienes/pharmacokinetics , Adolescent , Adult , Area Under Curve , Biotransformation , Cholestenone 5 alpha-Reductase , Free Radical Scavengers/pharmacology , Half-Life , Humans , Male , Neuroprotective Agents/pharmacology , Reference Values
16.
Drug Metab Dispos ; 26(10): 1048-51, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9763414

ABSTRACT

Bropirimine (2-amino-5-bromo-6-phenyl-4-pyrimidinone) is a member of a class of antineoplastic agents known as aryl pyrimidinones. In human liver microsomal incubations, bropirimine oxidative metabolism is characterized by the formation of three metabolites. Mass spectrometric analysis of the incubation mixture revealed three bropirimine oxidative metabolites, identified as the bropirimine dihydrodiol, p-hydroxybropirimine, and m-hydroxybropirimine. In vitro studies using human liver microsomes and recombinant cytochrome P450 isoforms were performed to identify the P450 enzyme(s) responsible for bropirimine oxidation. Coincubation with the selective CYP1A2 inhibitor alpha-naphthoflavone abolished bropirimine metabolism in human liver microsomes. Furthermore, when screened against a panel of cDNA expressed cytochrome P450 enzymes (CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4), bropirimine was metabolized to both p- and m-hydroxybropirimine exclusively in incubations with cDNA-expressed CYP1A2 microsomes. Mechanistic studies using cDNA-expressed CYP1A2 microsomes fortified with microsomal epoxide hydrolase revealed that all three bropirimine oxidative metabolites appear to be the result of a common arene oxide, which serves as a substrate for microsomal epoxide hydrolase to generate the dihydrodiol or rearranges to yield p- and m-hydroxybropirimine.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cytosine/analogs & derivatives , Biotransformation , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP1A2/metabolism , Cytosine/pharmacokinetics , Humans , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Oxidation-Reduction
17.
Xenobiotica ; 27(11): 1131-45, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9413917

ABSTRACT

1. We have determined that 2,4-dipyrrolidinylpyrimidine (2,4-DPP), used as a model for studies of the metabolism of therapeutic agents containing this moiety, undergoes three characteristic hydroxylations when incubated with male rat liver microsomes. Analysis of microsomal incubates of stable isotope labelled analogues of 2,4-DPP by particle beam-liquid chromatography-mass spectrometry (LC-PB-MS) has shown that the three metabolites are 4-(3-hydroxypyrrolidinyl)-2-(pyrrolidinyl)-pyrimidine (M1), 4-(2-hydroxypyrrolidinyl)-2-(pyrrolidinyl)-pyrimidine (M2) and 2-(2-hydroxypyrrolidinyl)-4-(pyrrolidinyl)-pyrimidine (M3). 2. We determined that enzymes of the cytochrome P450 family are responsible for the in vitro hydroxylations of 2,4-DPP. 3. We observed that in microsomal incubations carried out in the presence of cyanide, a single cyanide adduct is formed implicating an iminium ion intermediate in the oxidation of the 2-pyrrolidine ring. 4. We also determined the intermolecular deuterium isotope effects for the formation of each of the three products. For M1, kH/kD = 14.55 +/- 0.54; for M2, kH/kD = 6.01 +/- 0.65; and for M3, kH/kD = 5.35 +/- 1.18. 5. We interpret these data as suggesting that M2 and M3 are formed by the same mechanism, probably including the formation of an iminium ion, and that M1 is formed by direct hydrogen abstraction.


Subject(s)
Antioxidants/metabolism , Cytochrome P-450 Enzyme System/metabolism , Free Radical Scavengers , Pregnatrienes/metabolism , Pyrimidines/metabolism , Pyrroles/metabolism , Animals , Biotransformation , Chromatography, Liquid , Cyanides/metabolism , Deuterium , Hydroxylation , Male , Mass Spectrometry , Oxidation-Reduction , Pregnatrienes/pharmacokinetics , Rats , Rats, Sprague-Dawley
18.
Drug Metab Dispos ; 25(10): 1157-64, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9321519

ABSTRACT

Human hepatic microsomes were used to investigate the carboxylesterase-mediated bioactivation of CPT-11 to the active metabolite, SN-38. SN-38 formation velocity was determined by HPLC over a concentration range of 0.25-200 microM CPT-11. Biphasic Eadie Hofstee plots were observed in seven donors, suggesting that two isoforms catalyzed the reaction. Analysis by nonlinear least squares regression gave KM estimates of 129-164 microM with a Vmax of 5.3-17 pmol/mg/min for the low affinity isoform. The high affinity isoform had KM estimates of 1.4-3.9 microM with Vmax of 1.2-2.6 pmol/mg/min. The low KM carboxylesterase may be the main contributor to SN-38 formation at clinically relevant hepatic concentrations of CPT-11. Using standard incubation conditions, the effects of potential inhibitors of carboxylesterase-mediated CPT-11 hydrolysis were evaluated at concentrations >/= 21 microM. Positive controls bis-nitrophenylphosphate (BNPP) and physostigmine decreased CPT-11 hydrolysis to 1.3-3.3% and 23% of control values, respectively. Caffeine, acetylsalicylic acid, coumarin, cisplatin, ethanol, dexamethasone, 5-fluorouracil, loperamide, and prochlorperazine had no statistically significant effect on CPT-11 hydrolysis. Small decreases were observed with metoclopramide (91% of control), acetaminophen (93% of control), probenecid (87% of control), and fluoride (91% of control). Of the compounds tested above, based on these in vitro data, only the potent inhibitors of carboxylesterase (BNPP, physostigmine) have the potential to inhibit CPT-11 bioactivation if administered concurrently. The carboxylesterase-mediated hydrolysis of alpha-naphthyl acetate (alpha-NA) was used to determine whether CPT-11 was an inhibitor of hydrolysis of high turnover substrates of carboxylesterases. Inhibition of alpha-NA hydrolysis by CPT-11 was determined relative to positive controls BNPP and NaF. Incubation with microsomes pretreated with CPT-11 (80-440 microM) decreased alpha-naphthol formation to approximately 80% of control at alpha-NA concentrations of 50-800 microM. The inhibitors BNPP (360 microM) and NaF (500 microM) inhibited alpha-naphthol formation to 9-10% of control and to 14-20% of control, respectively. Therefore, CPT-11-sensitive carboxylesterase isoforms may account for only 20% of total alpha-NA hydrolases. Thus, CPT-11 is unlikely to significantly inhibit high turnover, nonselective substrates of carboxylesterases.


Subject(s)
Antineoplastic Agents, Phytogenic/metabolism , Camptothecin/analogs & derivatives , Carboxylic Ester Hydrolases/metabolism , Microsomes, Liver/metabolism , Biotransformation , Camptothecin/metabolism , Drug Interactions , Humans , Irinotecan
19.
Drug Metab Dispos ; 25(10): 1211-4, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9321526

ABSTRACT

The selectivity of inhibition for four dopamine receptor agonists (pramipexole, ropinirole, pergolide, and bromocriptine) on six human cytochrome P450 enzyme activities were evaluated using a simple in vitro inhibition screen. Drug-P450 interactions characterized as potent (i.e. greater than 50% inhibition of control enzyme activity) were then further examined to determine an IC50 for the interaction. Of the dopamine receptor agonists tested, three drugs, ropinirole, pergolide, and bromocriptine, were found to inhibit the activity of at least one human cytochrome P450 enzyme, while the remaining dopamine agonist, pramipexole, was devoid of any potent P450 interaction. None of the agonists tested inhibited the P450 marker activities of 2C9, 2C19, and 2E1. However, partial inhibition was observed between ropinirole and CYP1A2 and pergolide and CYP3A4. In contrast, potent interactions were observed between CYP2D6 and pergolide and ropinirole, as well as with CYP3A4 and bromocriptine. The results of this study indicate several drug P450 interactions; however, the likelihood of an in vivo interaction with these drugs remains to be established.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Dopamine Agonists/pharmacology , Enzyme Inhibitors/pharmacology , Benzothiazoles , Bromocriptine/pharmacology , Humans , Indoles/pharmacology , Pergolide/pharmacology , Pramipexole , Thiazoles/pharmacology
20.
J Pharmacol Exp Ther ; 277(2): 982-90, 1996 May.
Article in English | MEDLINE | ID: mdl-8627581

ABSTRACT

Tirilazad mesylate (Freedox), a potent inhibitor of membrane lipid peroxidation in vitro, is under clinical development for the treatment of subarachnoid hemorrhage. In humans, tirilazad is cleared almost exclusively via hepatic elimination. Characterization of three major microsomal metabolites of tirilazad by mass spectrometry indicated that hydroxylation had occurred in the pyrrolidine ring(s) and at the 6 beta-position of the steroid domain. A role for CYP3A4 in the formation of the three major metabolites (tirilazad hydroxylase activity) was established in human liver microsomal preparations: 1) Tirilazad hydroxylation was potently inhibited by troleandomycin and ketoconazole, specific inhibitors of CYP3A4. 2) The rates of tirilazad hydroxylation within a population of 14 human livers displayed a 9-fold interindividual variation and a significant correlation (r2 = .95) between tirilazad hydroxylation and testosterone 6 beta-hydroxylation. 3) Kinetic analysis of tirilazad hydroxylase activity in three human livers resulted in an apparent Km of 2.12, 1.68 and 1.66 microM, and Vmax = 0.85, 0.44 and 3.45 (nmol/mg protein/min) for HL14, HL17 and HL21, respectively. In addition, an apparent Km of 2.07 microM was established for tirilazad hydroxylation in a cDNA-expressed CYP3A4 microsomal system. Collectively, these data indicate that the metabolic clearance of tirilazad in humans is catalyzed primarily by CYP3A4 and provide an insight into factors (i.e., age, sex, drug-drug interactions) that modulate the metabolic clearance of tirilazad in vivo.


Subject(s)
Antioxidants/pharmacokinetics , Cytochrome P-450 Enzyme System/physiology , Microsomes, Liver/metabolism , Mixed Function Oxygenases/physiology , Pregnatrienes/pharmacokinetics , Biotransformation , Cytochrome P-450 CYP3A , Humans , Hydroxylation , Steroid Hydroxylases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...