Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Drug Metab Dispos ; 29(8): 1136-45, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11454733

ABSTRACT

Linezolid (Zyvox), the first of a new class of antibiotics, the oxazolidinones, is approved for treatment of Gram-positive bacterial infections, including resistant strains. The disposition of linezolid in human volunteers was determined, after a 500-mg (100-microCi) oral dose of [(14)C]linezolid. Radioactive linezolid was administered as a single dose, or at steady-state on day 4 of a 10-day, 500-mg b.i.d. regimen of unlabeled linezolid (n = 4/sex/regimen). Mean recovery of radioactivity in excreta was 93.8 +/- 1.1% (range 91.2-95.2%, n = 15), of which 83.9 +/- 3.3% (range 76.7-88.4%) was in urine and 9.9 +/- 3.4% (range 5.3-16.9%) was in feces. There was no major difference in rate or route of excretion of radioactivity by dose regimen. Linezolid was excreted primarily intact, and as two inactive, morpholine ring-oxidized metabolites, PNU-142586 and PNU-142300. Other minor metabolites were characterized by high-performance liquid chromatography-atmospheric pressure chemical ionization-mass spectrometry and (19)F NMR spectroscopy. After the single radioactive dose, linezolid was the major circulating drug-related material accounting for about 78% (male) and 93% (female) of the radioactivity area under the curve (AUC). PNU-142586 (T(max) of 3-5 h) accounted for about 26% (male) and 9% (female) of the radioactivity AUC. PNU-142300 (T(max) of 2-3 h) accounted for about 7% (male) and 4% (female) of the radioactivity AUC. Overall, mean linezolid and PNU-142586 exposures at steady-state were similar across sex. In conclusion, linezolid circulates in plasma mainly as parent drug. Linezolid and two major, inactive metabolites account for the major portion of linezolid disposition, with urinary excretion representing the major elimination route. Formation of PNU-142586 was the rate-limiting step in the clearance of linezolid.


Subject(s)
Acetamides/pharmacokinetics , Anti-Bacterial Agents/pharmacokinetics , Oxazolidinones/pharmacokinetics , Acetamides/blood , Acetamides/urine , Adult , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/urine , Biotransformation , Chromatography, High Pressure Liquid , Feces/chemistry , Female , Fluorine Radioisotopes , Half-Life , Humans , Isotope Labeling , Linezolid , Male , Mass Spectrometry , Middle Aged , Oxazolidinones/blood , Oxazolidinones/urine , Spectrophotometry, Ultraviolet , Whole-Body Counting
2.
Drug Metab Dispos ; 29(7): 1029-34, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11408370

ABSTRACT

Dapsone activates CYP2C9-mediated metabolism in various expression systems and is itself metabolized by CYP2C9 to its hydroxylamine metabolite. Studies were conducted with expressed CYP2C9 to characterize the kinetic effects of dapsone (0-100 microM) on (S)-flurbiprofen (2-300 microM), (S)-naproxen (10-1800 microM), and piroxicam (5-900 microM) metabolism in 6 x 6 matrix design experiments. The influence of (S)-flurbiprofen on dapsone hydroxylamine formation was also studied. Dapsone increased the Michaelis-Menten-derived V(max) of flurbiprofen 4'-hydroxylation from 12.6 to 20.6 pmol/min/pmol P450, and lowered its K(m) from 28.9 to 10.0 microM, suggesting that dapsone activates CYP2C9-mediated flurbiprofen metabolism without displacing flurbiprofen from the active site, supporting a two-site model describing activation. Similar results were observed with piroxicam 5'-hydroxylation, as V(max) was increased from 0.08 to 0.20 pmol/min/pmol P450 and K(m) was decreased from 183 to 50 microM in the presence of dapsone. In addition, the kinetic profile for naproxen was converted from biphasic to hyperbolic in the presence of dapsone, while exhibiting similar decreases in K(m) and increases in V(max). Kinetic parameters were also estimated using the two-site binding equation, with alpha values <1 and beta values >1, indicative of activation. Additionally, dapsone hydroxylamine formation was measured from incubations containing flurbiprofen, exhibiting a kinetic profile that was minimally affected by the presence of flurbiprofen. Overall, these results suggest that dapsone activates the metabolism of multiple substrates of CYP2C9 by binding within the active site and causing positive cooperativity, thus lending further support to a two-site binding model of P450-mediated metabolism.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Dapsone/pharmacology , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/metabolism , Enzyme Activation , Flurbiprofen/pharmacokinetics , Hydroxylation , Models, Chemical , Naproxen/pharmacokinetics , Piroxicam/pharmacokinetics , Substrate Specificity
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Drug Metab Dispos ; 26(7): 631-9, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9660845

ABSTRACT

The metabolism of delavirdine was examined using liver microsomes from several species with the aim of comparing metabolite formation among species and characterizing the enzymes responsible for delavirdine metabolism. Incubation of 10 microM [14C]delavirdine with either an S9 fraction from human jejunum or liver microsomes from rat, human, dog, or monkey followed by high pressure liquid chromatography analysis showed qualitatively similar metabolite profiles among species with the formation of three significant metabolites. The major metabolite was desalkyl delavirdine; however, the identity of MET-7 and MET-7a (defined by high pressure liquid chromatography elution) could not be unambiguously established, but they seem to be related pyridine hydroxy metabolites, most likely derived from 6'-hydroxylation of the pyridine ring. The apparent KM for delavirdine desalkylation activity ranged from 4.4 to 12.6 microM for human, rat, monkey, and dog microsomes, whereas Vmax ranged from 0.07 to 0.60 nmol/min/mg protein, resulting in a wide range of intrinsic clearance (6-135 microL/min/mg protein). Delavirdine desalkylation by microsomes pooled from several human livers was characterized by a KM of 6.8 +/- 0.8 microM and Vmax of 0. 44 +/- 0.01 nmol/min/mg. Delavirdine desalkylation among 23 human liver microsomal samples showed a meaningful correlation (r = 0.96) only with testosterone 6beta-hydroxylation, an indicator of CYP3A activity. Among ten human microsomal samples selected for uniform distribution of CYP3A activity, formation of MET-7 was strongly correlated with CYP3A activity (r = 0.95) and with delavirdine desalkylation (r = 0.98). Delavirdine desalkylation was catalyzed by cDNA-expressed CYP2D6 (KM 10.9 +/- 0.8 microM) and CYP3A4 (KM 5.4 +/- 1.4 microM); however, only CYP3A4 catalyzed formation of MET-7 and MET-7a. Quinidine inhibited human liver microsomal delavirdine desalkylation by about 20%, indicating a minor role of CYP2D6. These findings suggest the potential for clinical interaction with coadministered drugs that are metabolized by or influence the activity of CYP3A or CYP2D6.


Subject(s)
Anti-HIV Agents/metabolism , Cytochrome P-450 CYP2D6/physiology , Cytochrome P-450 Enzyme System/physiology , Delavirdine/metabolism , HIV-1/drug effects , Microsomes, Liver/metabolism , Mixed Function Oxygenases/physiology , Reverse Transcriptase Inhibitors/metabolism , Animals , Cytochrome P-450 CYP3A , Dogs , Female , Humans , Macaca fascicularis , Male , Mass Spectrometry , Models, Molecular , Rats , Rats, Sprague-Dawley
9.
J Chromatogr B Biomed Sci Appl ; 705(1): 39-45, 1998 Jan 23.
Article in English | MEDLINE | ID: mdl-9498668

ABSTRACT

We report here a quantitative methodology developed for determination of SEPA (2-n-nonyl-1,3-dioxolane) in human serum. The method employed solid-phase extraction of SEPA and internal standard, [13C2]SEPA, from serum followed by gas chromatography-mass spectrometry analysis using EI monitoring m/z 73 and 75. We have investigated the utility of stable isotope dilution gas chromatography-mass spectrometry (GC-MS) for the determination of SEPA concentrations in serum using chemical ionization (positive ion, CI) or electron ionization (EI). The comparison of the specificity and sensitivity between EI and CI indicated that monitoring the m/z 73 ion in EI was superior to monitoring either MH+ or m/z 73 using CI. The method was simple, sensitive and accurate, demonstrating a lower limit of quantitation (LLOQ) of 0.25 ng/ml and intra- and inter-assay accuracy and precision of < or = 7.5%.


Subject(s)
Adjuvants, Pharmaceutic/analysis , Dioxolanes/blood , Adjuvants, Pharmaceutic/administration & dosage , Administration, Topical , Carbon Isotopes , Gas Chromatography-Mass Spectrometry , Humans , Indicator Dilution Techniques , Minoxidil/administration & dosage , Minoxidil/blood , Reproducibility of Results , Sensitivity and Specificity
10.
Xenobiotica ; 27(10): 1039-51, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9364741

ABSTRACT

1. Metabolites of the cyclic bisphosphonate ester, 4-[2,2'-bis-(5,5- dimethyl-1,3,2-dioxaphosphorinan-2-yl)] butanoyl-3-fluoro-benzene (PNU-91638) in bile or urine of the male Sprague-Dawley rat were characterized by mass spectrometry. The chronically bile duct/duodenal-cannulated male rats received a single oral dose of 100 mg/kg [13C] [14C]PNU-91638. Bile and urine samples were analysed for radioactivity and profiled by hplc with radiometric and UV detection. 2. The 0-28-h bile and urine accounted for 46.0 and 19.7% of dose respectively. The structures of radioactive peaks were investigated by ionspray and liquid secondary ion mass spectrometry (LSIMS) and LSIMS/MS analysis. 3. Major metabolites in urine included two regioisomeric phenol glucuronides, a gem methyl hydroxylated metabolite of the bisphosphonate heterocycle, a phenol metabolite, parent drug and a benzylic alcohol metabolite. Additional metabolites in bile included an unstable phenol/glutathione adduct (from a putative epoxide intermediate) with several minor isobaric regioisomers, and a carboxylic acid derived from the gem methyl hydroxylated bisphosphonate ring. 4. The structures proposed have not been confirmed by nmr due to discontinuation of the development of PNU-91638.


Subject(s)
Antirheumatic Agents/metabolism , Diphosphonates/metabolism , Animals , Antirheumatic Agents/pharmacokinetics , Antirheumatic Agents/urine , Bile/metabolism , Biliary Tract/metabolism , Carbon Radioisotopes , Chromatography, Liquid , Diphosphonates/pharmacokinetics , Diphosphonates/urine , Drug Stability , Male , Mass Spectrometry , Rats , Rats, Sprague-Dawley
11.
Drug Metab Dispos ; 25(7): 828-39, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9224777

ABSTRACT

Delavirdine mesylate (U-90152T) is a highly specific nonnucleoside HIV-1 reverse transcriptase inhibitor currently under development for the treatment of AIDS. The excretion, disposition, brain penetration, and metabolism of delavirdine were investigated in CD-1 mice after oral administration of [14C]delavirdine mesylate at single doses of 10 and/or 250 mg/kg and multiple doses of 200 mg/kg/day. Studies were conducted with 14C-carboxamide and 2-14C-pyridine labels, as well as 13C3-labeled drug to facilitate metabolite identification. Excretion was dose dependent with 57-70% of the radioactivity eliminated in feces and 25-36% in urine. Pharmacokinetic analyses of delavirdine and its N-desisopropyl metabolite (desalkyl delavirdine) in plasma showed that delavirdine was absorbed and metabolized rapidly, that it constituted a minor component in circulation, that its pharmacokinetics were nonlinear, and that its metabolism to desalkyl delavirdine was capacity limited or inhibitable. Delavirdine did not significantly cross the blood-brain barrier; however, its N-isopropylpyridinepiperazine metabolite arising from amide bond cleavage-was present in brain at levels 2- to 3-fold higher than in plasma. The metabolism of delavirdine in the mouse was extensive and involved amide bond cleavage, N-desalkylation, hydroxylation at the C-6' position of the pyridine ring, and pyridine ring-cleavage as determined by MS and/or 1H and 13C NMR spectroscopies. N-desalkylation and amide bond cleavage were the primary metabolic pathways at low drug doses and, as the biotransformation of delavirdine to desalkyl delavirdine reached saturation or inhibition, amide bond cleavage became the predominant pathway at higher doses and after multiple doses.


Subject(s)
Anti-HIV Agents/pharmacokinetics , HIV-1/enzymology , Indoles/pharmacokinetics , Piperazines/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Animals , Biotransformation , Chromatography, High Pressure Liquid , Delavirdine , Feces/chemistry , Female , Hydrolysis , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Mice , Spectrophotometry, Ultraviolet , Tissue Distribution
12.
Drug Metab Dispos ; 25(7): 814-27, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9224776

ABSTRACT

Delavirdine mesylate (U-90152T) is a highly specific nonnucleoside HIV-1 reverse transcriptase inhibitor currently under development for the treatment of AIDS. The metabolism of delavirdine was investigated in male and female cynomolgus monkeys after oral administration of [14C-carboxamide]delavirdine mesylate at single doses of 80 mg/kg and multiple doses of 160 to 300 mg/kg/day. Desalkyl delavirdine was the major metabolite in circulation. In urine, desalkyl delavirdine accounted for nearly half of the radioactivity, with despyridinyl delavirdine and conjugates of desalkyl delavirdine accounting for most of the remaining radioactivity. Bile was mostly composed of desalkyl delavirdine and 6'-O-glucuronide delavirdine, with parent drug, 4-O-glucuronide delavirdine, and conjugates of desalkyl delavirdine as significant components. In addition, several minor metabolites were observed in urine and bile of delavirdine treated monkeys. The metabolism of delavirdine in the monkey was extensive and involved N-desalkylation, hydroxylation at the C-4' and C-6' positions of the pyridine ring, hydroxylation at the C-4 position of the indole ring, pyridine ring-cleavage, N-glucuronidation of the indole ring, and amide bond cleavage as determined by MS and/or one-dimensional and two-dimensional NMR spectroscopies. Phase II biotransformations included glucuronidation, sulfation, and beta-N-acetylglucosaminidation. The identification of the N-linked beta-N-acetylglucosamine and 4-O-glucuronide metabolites of delavirdine represents novel biotransformation pathways.


Subject(s)
Anti-HIV Agents/pharmacokinetics , HIV-1/enzymology , Indoles/pharmacokinetics , Piperazines/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Animals , Bile/chemistry , Bile/metabolism , Biotransformation , Chromatography, High Pressure Liquid , Delavirdine , Female , Hydrolysis , Macaca fascicularis , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Spectrophotometry, Ultraviolet
13.
Drug Metab Dispos ; 25(2): 228-42, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9029054

ABSTRACT

Delavirdine mesylate (U-90152T) is a highly specific nonnucleoside reverse transcriptase inhibitor currently under development for the treatment of AIDS. The excretion, disposition, and metabolism of delavirdine were investigated in Sprague-Dawley rats after oral administration of [14C]delavirdine mesylate at single doses ranging from 10 to 250 mg/kg and multiple doses ranging from 20 to 250 mg/kg/day. Excretion studies showed that feces was the major route of elimination, delavirdine was well absorbed (>80%) after a 10 mg/kg single dose, and excretion was dose-dependent. The metabolism of delavirdine in the rat was extensive. The following metabolites were identified (% of dose in rats given 10 and 100 mg/kg, respectively): 6'-hydroxy delavirdine (7.1% and 15.6%) and its glucuronide (12.2% and 6.2%) and sulfate (5.5% and 3.2%) conjugates, despyridinyl delavirdine (12.1% and 11.7%) and its conjugate (13.0% and 11.7%), desalkyl delavirdine (16.5% and 13.4%), and its N-sulfamate, 6'- and 4'-sulfate conjugates (2.9% and 3.9%). Cleavage of the amide bond in delavirdine to give N-isopropylpyridinepiperazine and indole carboxylic acid constituted a minor pathway. Degradation of 6'-hydroxy delavirdine generated despyridinyl delavirdine and the pyridine-ring opened MET-14. The metabolic pathway of delavirdine involved N-desalkylation, pyridine ring hydroxylation, pyridine ring cleavage, and amide bond cleavage.


Subject(s)
Anti-HIV Agents/pharmacokinetics , HIV-1/drug effects , Indoles/pharmacokinetics , Piperazines/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Administration, Oral , Animals , Bile/chemistry , Chromatography, High Pressure Liquid , Delavirdine , Feces/chemistry , Female , HIV-1/enzymology , Indoles/urine , Male , Mass Spectrometry , Piperazines/urine , Rats , Rats, Sprague-Dawley
14.
Chem Res Toxicol ; 9(8): 1230-9, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8951224

ABSTRACT

U-89843 is a novel pyrrolo[2,3-d]pyrimidine antioxidant with prophylactic activity in animal models of lung inflammation. During preclinical safety evaluation, U-89843 was found to give a positive response in the in vitro unscheduled DNA synthesis (UDS) assay, an assay which measures DNA repair following chemically-induced DNA damage in metabolically competent rat hepatocytes. Incubation of [14C]U-89843 with liver microsomes resulted in covalent binding of radioactive material to macromolecules by a process that was NADPH-dependent. U-89843 has been shown to undergo C-6 methylhydroxylation to give U-97924, in rat both in vivo and in vitro, in a reaction catalyzed by cytochrome P450 2C11. Synthetical U-97924 is chemically reactive and undergoes dimerization in aqueous solution. The dimerization of U-97924 was significantly inhibited by addition of nucleophiles such as methanol, glutathione, and N-acetylcysteine. Characterization of the corresponding methanol, glutathione, and N-acetylcysteine adducts of U-97924 supported the hypothesis of a reaction pathway involving reactive iminium species formed via dehydration of U-97924. The metabolism-dependent irreversible covalent binding of radioactive material to liver microsomal protein and DNA also is dramatically reduced in the presence of reduced glutathione (GSH). A trifluoromethyl analog of U-89843 was prepared in an effort to block the corresponding metabolic hydroxylation pathway. This new compound (U-107634) was found to be negative in the in vitro UDS assay, and its metabolic susceptibility toward hydroxylation at the C-6 methyl group was eliminated. These observations suggest that the positive in vitro UDS results of U-89843 are mediated by the bioactivation of U-89843, leading to reactive electrophilic intermediates derived from the (hydroxymethyl)pyrrole metabolite U-97924.


Subject(s)
Free Radical Scavengers/toxicity , Mutagens/toxicity , Pyrimidines/toxicity , Pyrroles/toxicity , Animals , Biotransformation , Chromatography, High Pressure Liquid , DNA Repair , Dogs , Free Radical Scavengers/chemistry , Free Radical Scavengers/metabolism , Gas Chromatography-Mass Spectrometry , Haplorhini , Humans , In Vitro Techniques , Male , Microsomes, Liver/metabolism , Mutagens/chemistry , Mutagens/metabolism , Protein Binding , Pyrimidines/chemistry , Pyrimidines/metabolism , Pyrroles/chemistry , Pyrroles/metabolism , Rats , Rats, Inbred F344 , Spectrometry, Mass, Fast Atom Bombardment , Spectrophotometry, Ultraviolet
15.
Drug Metab Dispos ; 24(2): 187-98, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8742230

ABSTRACT

The biotransformation of 6,7-dimethyl-2,4-di-1-pyrrolidinyl-7H-pyrrolo[2,3-d]pyrimidine (U-89843) has been studied in rat both in vitro and in vivo. Major metabolites observed by HPLC analysis of rat plasma, liver cytosol, and microsomal incubations were characterized by UV, LC/MS, and comparison with synthetic standards. The structures of the metabolites were shown to be the C-6 hydroxymethyl (U-97924), C-6 formyl (U-97865), and C-6 carboxyl analogs of U-89843. In the male rat, formation of U-97924 is mediated by cytochrome P4502C11. Kinetic analysis of U-97924 formation indicated that it was a high-affinity/high-capacity process (KM = 4.2 +/- 0.5 microM; Vmax = 21.2 +/- 0.8 nmol/mg/min). Formation of U-97865 via enzymatic oxidation from the primary metabolite U-97924 was catalyzed by both the microsomal subcellular fraction in a NADPH-dependent process (presumably via cytochrome P450) and in cytosol by NAD(+)-dependent alcohol dehydrogenase. Upon incubation with cytosolic fractions, U-97865 was found to undergo NAD(+)-dependent oxidation, mediated by aldehyde dehydrogenase, to the corresponding carboxylic acid. Although significant levels of U-89843, U-97924, and U-97865 were observed in vivo in rat plasma, only a minor amount of the carboxylic acid together with larger amounts of unidentified polar metabolites were excreted in urine and feces.


Subject(s)
Antioxidants/pharmacokinetics , Microsomes, Liver/metabolism , Pyrimidines/pharmacokinetics , Pyrroles/pharmacokinetics , Administration, Oral , Animals , Biotransformation , Chromatography, High Pressure Liquid , Gas Chromatography-Mass Spectrometry , Male , Molecular Structure , Rats , Rats, Sprague-Dawley , Spectrophotometry, Ultraviolet
16.
Drug Metab Dispos ; 24(1): 65-73, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8825192

ABSTRACT

The major metabolites of the bisphosphonate ester U-91502 were isolated from the bile and urine of male Sprague-Dawley rats and identified by NMR and MS. Bile duct-exteriorized and taurocholate-supplemented rats received single oral doses of 10-140 mg/kg of labeled U-91502. Analysis of radioactivity in bile, urine, and feces showed that U-91502-related radioactivity was rapidly excreted, predominantly in bile, achieving peak concentrations in bile of 1250 +/- 622 micrograms-eq/g during first 3 hr after a 10 mg/kg dose. The three major drug-related materials in bile and urine were separated by HPLC and designated in order of reversed-phase elution as metabolites A, B, and C. The least polar metabolite (C) was shown by HPLC/particle beam/MS and HPLC/electrospray/MS to be the triester, U-94532. Metabolite C cochromatographed with a synthesized standard of U-94532A. Metabolite B was the glucuronide conjugate of the 5-hydroxy pyrimidinone, U-97294. Metabolite A was a product of glutathione addition to a putative pyrimidinone 4,5-epoxide. Mechanisms for the formation of metabolites A, B, and C based on metabolite structure and stability were proposed.


Subject(s)
Diphosphonates/pharmacokinetics , Organophosphorus Compounds/pharmacokinetics , Animals , Bile/metabolism , Biological Availability , Chromatography, High Pressure Liquid , Diphosphonates/metabolism , Diphosphonates/urine , Male , Mass Spectrometry/methods , Organophosphorus Compounds/metabolism , Organophosphorus Compounds/urine , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL