Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 29(12): 1614-28, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11717181

ABSTRACT

Rofecoxib is a potent and highly selective cyclooxygenase-2 inhibitor used for the treatment of osteoarthritis and pain. Following administration of [4-(14)C]rofecoxib to intact rats, the plasma C(max) (at approximately 1 h) was followed by a secondary C(max) (at approximately 10 h), which was not observed in bile duct-cannulated rats. Following administration of [4-(14)C]5-hydroxyrofecoxib to intact or bile duct-cannulated rats, radiolabeled rofecoxib was detected in plasma, and once again a secondary C(max) for rofecoxib was observed (at approximately 10 h), which occurred only in the intact animals. These results indicate that reversible metabolism of rofecoxib to 5-hydroxyrofecoxib occurs in the rat and that the process is dependent upon an uninterrupted bile flow. Studies on the contents of the gastrointestinal tract of rats showed that conversion of 5-hydroxyrofecoxib to parent compound occurs largely in the lower intestine. Treatment of rats with [5-(18)O]5-hydroxyrofecoxib, followed by liquid chromatography-tandem mass spectrometry analyses of plasma samples, confirmed that 5-hydroxyrofecoxib undergoes metabolism to the parent drug, yielding [1-(18)O]rofecoxib, [2-(18)O]rofecoxib, and unlabeled rofecoxib. Similarly, treatment with [1,2-(18)O(2)]rofecoxib afforded the same three isotopic variants of rofecoxib. These findings are consistent with a metabolic sequence involving 5-hydroxylation of rofecoxib, biliary elimination of the corresponding glucuronide, and deconjugation of the glucuronide in the lower gastrointestinal tract. Reduction of the 5-hydroxyrofecoxib thus liberated yields a hydroxyacid that cyclizes spontaneously to regenerate rofecoxib, which is reabsorbed and enters the systemic circulation. This sequence represents a novel form of enterohepatic recycling and reflects the susceptibility of 5-hydroxyrofecoxib, as well as rofecoxib itself, to reversible 2-furanone ring opening under in vivo conditions.


Subject(s)
Cyclooxygenase Inhibitors/pharmacokinetics , Lactones/metabolism , Lactones/pharmacokinetics , Animals , Bile/metabolism , Biotransformation , Carbon Radioisotopes , Chromatography, High Pressure Liquid , Furans/metabolism , Intestinal Absorption , Isotope Labeling , Magnetic Resonance Spectroscopy , Male , Oxygen Isotopes , Rats , Rats, Sprague-Dawley , Sulfones , Tissue Distribution
2.
Br J Clin Pharmacol ; 52(1): 69-76, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11453892

ABSTRACT

AIMS: Patients with migraine may receive the 5-HT1B/1D agonist, rizatriptan (5 or 10 mg), to control acute attacks. Patients with frequent attacks may also receive propranolol or other beta-adrenoceptor antagonists for migraine prophylaxis. The present studies investigated the potential for pharmacokinetic or pharmacodynamic interaction between beta-adrenoceptor blockers and rizatriptan. METHODS: Four double-blind, placebo-controlled, randomized crossover investigations were performed in a total of 51 healthy subjects. A single 10 mg dose of rizatriptan was administered after 7 days' administration of propranolol (60 and 120 mg twice daily), nadolol (80 mg twice daily), metoprolol (100 mg twice daily) or placebo. Rizatriptan pharmacokinetics were assessed. In vitro incubations of rizatriptan and sumatriptan with various beta-adrenoceptor blockers were performed in human S9 fraction. Production of the indole-acetic acid-MAO-A metabolite of each triptan was measured. RESULTS: Administration of rizatriptan during propranolol treatment (120 mg twice daily for 7.5 days) increased the AUC(0, infinity) for rizatriptan by approximately 67% and the Cmax by approximately 75%. A reduction in the dose of propranolol (60 mg twice daily) and/or the incorporation of a delay (1 or 2 h) between propranolol and rizatriptan administration did not produce a statistically significant change in the effect of propranolol on rizatriptan pharmacokinetics. Administration of rizatriptan together with nadolol (80 mg twice daily) or metoprolol (100 mg twice daily) for 7 days did not significantly alter the pharmacokinetics of rizatriptan. No untoward adverse experiences attributable to the pharmacokinetic interaction between propranolol and rizatriptan were observed, and no subjects developed serious clinical, laboratory, or other significant adverse experiences during coadministration of rizatriptan with any of the beta-adrenoceptor blockers. In vitro incubations showed that propranolol, but not other beta-adrenoceptor blockers significantly inhibited the production of the indole-acetic acid metabolite of rizatriptan and sumatriptan. CONCLUSIONS: These results suggest that propranolol increases plasma concentrations of rizatriptan by inhibiting monoamine oxidase-A. When prescribing rizatriptan to migraine patients receiving propranolol for prophylaxis, the 5 mg dose of rizatriptan is recommended. Administration with other beta-adrenoceptor blockers does not require consideration of a dose adjustment.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Metoprolol/pharmacology , Nadolol/pharmacology , Propranolol/pharmacology , Serotonin Receptor Agonists/pharmacokinetics , Triazoles/pharmacokinetics , Adolescent , Adult , Biological Availability , Cardiovascular System/drug effects , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , In Vitro Techniques , Male , Middle Aged , Receptor, Serotonin, 5-HT1B , Receptor, Serotonin, 5-HT1D , Receptors, Serotonin/metabolism , Tryptamines
3.
Drug Metab Dispos ; 28(10): 1244-54, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10997947

ABSTRACT

Absorption, distribution, metabolism, and excretion studies were conducted in rats and dogs with rofecoxib (VIOXX, MK-0966), a potent and highly selective inhibitor of cyclooxygenase-2 (COX-2). In rats, the nonexponential decay during the terminal phase (4- to 10-h time interval) of rofecoxib plasma concentration versus time curves after i.v. or oral administration of [(14)C]rofecoxib precluded accurate determinations of half-life, AUC(0-infinity) (area under the plasma concentration versus time curve extrapolated to infinity), and hence, bioavailability. After i.v. administration of [(14)C]rofecoxib to dogs, plasma clearance, volume of distribution at steady state, and elimination half-life values of rofecoxib were 3.6 ml/min/kg, 1.0 l/kg, and 2.6 h, respectively. Oral absorption (5 mg/kg) was rapid in both species with C(max) occurring by 0.5 h (rats) and 1.5 h (dogs). Bioavailability in dogs was 26%. Systemic exposure increased with increasing dosage in rats and dogs after i.v. (1, 2, and 4 mg/kg), or oral (2, 5, and 10 mg/kg) administration, except in rats where no additional increase was observed between the 5 and 10 mg/kg doses. Radioactivity distributed rapidly to tissues, with the highest concentrations of the i.v. dose observed in most tissues by 5 min and by 30 min in liver, skin, fat, prostate, and bladder. Excretion occurred primarily by the biliary route in rats and dogs, except after i.v. administration of [(14)C]rofecoxib to dogs, where excretion was divided between biliary and renal routes. Metabolism of rofecoxib was extensive. 5-Hydroxyrofecoxib-O-beta-D-glucuronide was the major metabolite excreted by rats in urine and bile. 5-Hydroxyrofecoxib, rofecoxib-3',4'-dihydrodiol, and 4'-hydroxyrofecoxib sulfate were less abundant, whereas cis- and trans-3,4-dihydro-rofecoxib were minor. Major metabolites in dog were 5-hydroxyrofecoxib-O-beta-D-glucuronide (urine), trans-3, 4-dihydro-rofecoxib (urine), and 5-hydroxyrofecoxib (bile).


Subject(s)
Cyclooxygenase Inhibitors/pharmacokinetics , Lactones/pharmacokinetics , Absorption , Animals , Area Under Curve , Bile/chemistry , Bile/metabolism , Carbon Radioisotopes , Chromatography, High Pressure Liquid , Cyclooxygenase Inhibitors/blood , Cyclooxygenase Inhibitors/metabolism , Dogs , Dose-Response Relationship, Drug , Kinetics , Lactones/metabolism , Lactones/urine , Male , Metabolic Clearance Rate , Rats , Rats, Sprague-Dawley , Species Specificity , Sulfones , Tissue Distribution
4.
Drug Metab Dispos ; 28(6): 633-42, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10820134

ABSTRACT

3-([4-(4-Chlorophenyl)piperazin-1-yl]-methyl)-1H-pyrrolo-2, 3-beta-pyridine (L-745,870) is a dopamine D(4) selective antagonist that has been studied as a potential treatment for schizophrenia, with the expectation that it would not exhibit the extrapyramidal side effects often observed with the use of classical antipsychotic agents. The metabolism of L-745,870 in vivo was investigated in the rat, rhesus monkey, and human using liquid chromatography-tandem mass spectrometry and/or NMR techniques in conjunction with radiochemical detection. In all three species, two major metabolic pathways were identified, namely N-dealkylation at the substituted piperazine moiety and the formation of a novel mercapturic acid adduct. It is proposed that the latter biotransformation process involves the formation of an electrophilic imine methide intermediate, analogous to that produced from 3-methyl indole. This report appears to represent the first example of metabolic activation of a 3-alkyl-7-azaindole nucleus.


Subject(s)
Acetylcysteine/urine , Dopamine Antagonists/metabolism , Pyridines/metabolism , Pyrroles/metabolism , Receptors, Dopamine D2/metabolism , Acetylcysteine/metabolism , Animals , Dopamine Antagonists/pharmacology , Dopamine Antagonists/urine , Dopamine D2 Receptor Antagonists , Humans , Macaca mulatta , Male , Pyridines/pharmacology , Pyridines/urine , Pyrroles/pharmacology , Pyrroles/urine , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D4
5.
Bioorg Med Chem Lett ; 10(2): 175-8, 2000 Jan 17.
Article in English | MEDLINE | ID: mdl-10673105

ABSTRACT

Furo[3,4-d]pyrimidinones were found to be metabolites of dihydropyrimidinones such as 1a-b that are subtype-selective antagonists of the alpha1a-adrenergic receptor. A versatile synthesis that provides access to furo[3,4-d]pyrimidinones in high yield and in enantiomerically pure forms is described along with structure-activity relationships in the series.


Subject(s)
Adrenergic alpha-Antagonists/chemical synthesis , Pyrimidinones/chemical synthesis , Receptors, Adrenergic, alpha-1/drug effects , Adrenergic alpha-Antagonists/pharmacology , Animals , Binding, Competitive , Molecular Structure , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Prazosin/metabolism , Pyrimidinones/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship
6.
Xenobiotica ; 30(1): 47-59, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10659950

ABSTRACT

1. Quantitative species differences and human liver enzymes involved in the metabolism of L-775,606, a potent and selective 5-HT1D receptor agonist developed for the acute treatment of migraine headache, have been investigated in vitro. 2. In human, monkey, dog and rat liver microsomes, formation of the hydroxylated M1 and the N-dealkylated M2 was mediated by enzyme(s) of high-affinity (apparent Km approximately 1-6 microM), and that of the two N-oxide isomers (M3) was catalysed by those of low affinity (apparent Km approximately 50-110 microM). In dog, M3 constituted a major pathway (approximately 40%), whereas in all other species it was a minor metabolite (< 5%). 3. In human liver microsomes, a marked inhibition (> or =80%) of M1 and M2 formation was observed by SKF525-A, troleandomycin, ketoconazole and anti-CYP3A antibodies, whereas the inhibition was modest (approximately 20-40%) with quercetin. Of seven cDNA-expressed human P450 tested, only CYP3A4 and CYP2C8 were capable of oxidizing L-775,606, resulting primarily in M1 and M2. However, CYP3A4 possessed much higher affinity (> or = 20-fold) and much higher intrinsic activity (> 100-fold) than CYP2C8. 4. In contrast, N-oxidation was not inhibited by any inhibitors of P450 tested, but rather was reduced significantly by heat treatment and methimazole, and was increased substantially with an incubation pH>7.4. Human flavin-containing monooxygenase form 3 (FMO3) catalysed exclusively the N-oxidation to M3, with apparent Km and optimum pH comparable with those observed in human liver microsomes. 5. These results demonstrated quantitative interspecies differences in the metabolism of L-775,606. In human, metabolism of L-775,606 to the principal metabolites, M1 and M2, was mediated primarily by CYP3A4 with minimal contribution from CYP2C8, whereas the minor N-oxidative pathway was catalysed mainly by FMO3.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Indoles/pharmacokinetics , Liver/metabolism , Monoamine Oxidase/metabolism , Piperazines/pharmacokinetics , Receptors, Serotonin/drug effects , Serotonin Receptor Agonists/pharmacokinetics , Animals , Antibodies, Blocking/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/biosynthesis , DNA, Complementary/biosynthesis , Dogs , Enzyme Inhibitors/pharmacology , Haplorhini , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Isoenzymes/biosynthesis , Isoenzymes/metabolism , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Rats , Receptor, Serotonin, 5-HT1D , Recombinant Proteins/metabolism , Species Specificity
7.
J Clin Pharmacol ; 40(1): 74-83, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10631625

ABSTRACT

Rizatriptan is a novel 5-HT1D/1B agonist for relief of migraine headache. The pharmacokinetics, metabolite profiles, and tolerability of rizatriptan were examined in a multiple-dose study in healthy subjects. Rizatriptan (N = 24) (or placebo, N = 12) was administered as a single 10 mg dose, followed 48 hours later by administration of one 10 mg dose every 2 hours for three doses on 4 consecutive days, corresponding to the maximum daily dose for a migraine attack. The AUC of rizatriptan and its active N-monodesmethyl metabolite after three 10 mg doses was approximately threefold greater than the plasma concentrations following a single 10 mg dose. Metabolite profiles were similar after single and multiple doses. Adverse events during rizatriptan were mild and transient; similar events occurred during placebo, with a somewhat reduced incidence. Diastolic blood pressure tended to increase compared with placebo (approximately 5 mmHg), particularly on the first multiple-dose day (p < .01 vs. placebo). In conclusion, rizatriptan is well tolerated by healthy subjects during multiple-dose administration, with no unexpected accumulation of drug in plasma.


Subject(s)
Serotonin Receptor Agonists/adverse effects , Serotonin Receptor Agonists/pharmacokinetics , Triazoles/adverse effects , Triazoles/pharmacokinetics , Adult , Blood Pressure/drug effects , Double-Blind Method , Female , Humans , Male , Middle Aged , Placebos , Serotonin Receptor Agonists/administration & dosage , Time Factors , Triazoles/administration & dosage , Tryptamines
8.
Drug Metab Dispos ; 28(1): 89-95, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10611145

ABSTRACT

The absorption and disposition of rizatriptan (MK-0462, Maxalt(TM)), a selective 5-HT(1B/1D) receptor agonist used in the treatment of migraine headaches, was investigated in humans. In a two-period, single i.v. (3 mg, 30-min infusion), and single oral (10 mg) dose study with [(14)C]rizatriptan in six healthy human males, total recovery of radioactivity was approximately 94%, with unchanged rizatriptan and its metabolites being excreted mainly in the urine (89% i.v. dose, 82% p.o. dose). Approximately 26 and 14% of i.v. and oral rizatriptan doses, respectively, were excreted in urine as intact parent drug. In a second, high-dose study (60 mg p.o.), five metabolites excreted into urine were identified using liquid chromatography-tandem mass spectrometry and NMR methods. They were triazolomethyl-indole-3-acetic acid, rizatriptan-N(10)-oxide, 6-hydroxy-rizatriptan, 6-hydroxy-rizatriptan sulfate, and N(10)-monodesmethyl-rizatriptan. Urinary excretion of triazolomethyl-indole-3-acetic acid after i.v. and oral administrations of rizatriptan accounted for 35 and 51% of the dose, respectively, whereas the corresponding values for rizatriptan-N(10)-oxide were 4 and 2% of the dose. Plasma clearance (CL) and renal clearance (CL(r)) were 1325 and 349 ml/min, respectively, after i.v. administration. A similar CL(r) value was obtained after oral administration (396 ml/min). The primary route of rizatriptan elimination occurred via nonrenal route(s) (i.e., metabolism) because the CL(r) of rizatriptan accounted for 25% of total CL. Furthermore, the CL(r) was higher than normal glomerular filtration rate ( approximately 130 ml/min), indicating that this compound was actively secreted by renal tubules. The absorption of rizatriptan was approximately 90%, but it experienced a moderate first-pass effect, resulting in a bioavailability estimate of 47%.


Subject(s)
Serotonin Receptor Agonists/pharmacokinetics , Triazoles/pharmacokinetics , Administration, Oral , Area Under Curve , Carbon Radioisotopes/metabolism , Chromatography, High Pressure Liquid , Cross-Over Studies , Feces , Humans , Infusions, Intravenous , Male , Migraine Disorders/drug therapy , Reference Values , Serotonin Receptor Agonists/administration & dosage , Serotonin Receptor Agonists/therapeutic use , Serotonin Receptor Agonists/urine , Tissue Distribution , Triazoles/administration & dosage , Triazoles/therapeutic use , Triazoles/urine , Tryptamines
9.
J Med Chem ; 42(23): 4778-93, 1999 Nov 18.
Article in English | MEDLINE | ID: mdl-10579841

ABSTRACT

We have previously described compound 1a as a high-affinity subtype selective alpha(1a) antagonist. In vitro and in vivo evaluation of compound 1a showed its major metabolite to be a mu-opioid agonist, 4-methoxycarbonyl-4-phenylpiperidine (3). Several dihydropyrimidinone analogues were synthesized with the goal of either minimizing the formation of 3 by modification of the linker or finding alternative piperidine moieties which when cleaved as a consequence of metabolism would not give rise to mu-opioid activity. Modification of the linker gave several compounds with good alpha(1a) binding affinity (K(i) = < 1 nM) and selectivity (>300-fold over alpha(1b) and alpha(1d)). In vitro analysis in the microsomal assay revealed these modifications did not significantly affect N-dealkylation and the formation of the piperidine 3. The second approach, however, yielded several piperidine replacements for 3, which did not show significant mu-opioid activity. Several of these compounds maintained good affinity at the alpha(1a) adrenoceptor and selectivity over alpha(1b) and alpha(1d). For example, the piperidine fragments of (+)-73 and (+)-83, viz. 4-cyano-4-phenylpiperidine and 4-methyl-4-phenylpiperidine, were essentially inactive at the mu-opioid receptor (IC(50) > 30 microM vs 3 microM for 3). Compounds (+)-73 and (+)-83 were subjected to detailed in vitro and in vivo characterization. Both these compounds, in addition to their excellent selectivity (>880-fold) over alpha(1b) and alpha(1d), also showed good selectivity over several other recombinant human G-protein coupled receptors. Compounds (+)-73 and (+)-83 showed good functional potency in isolated human prostate tissues, with K(b)s comparable to their in vitro alpha(1a) binding data. In addition, compound (+)-73 also exhibited good uroselectivity (DBP K(b)/IUP K(b) > 20-fold) in the in vivo experiments in dogs, similar to 1a.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-Antagonists/chemical synthesis , Piperidines/chemical synthesis , Pyrimidinones/chemical synthesis , Adrenergic alpha-Antagonists/chemistry , Adrenergic alpha-Antagonists/metabolism , Adrenergic alpha-Antagonists/pharmacology , Animals , Biological Availability , Blood Pressure/drug effects , Dogs , Drug Design , Drug Evaluation, Preclinical , GTP-Binding Proteins/metabolism , Half-Life , Humans , In Vitro Techniques , Male , Microsomes/metabolism , Piperidines/chemistry , Piperidines/metabolism , Piperidines/pharmacology , Prostate/metabolism , Pyrimidinones/chemistry , Pyrimidinones/metabolism , Pyrimidinones/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Opioid, mu/agonists , Recombinant Proteins/metabolism , Stereoisomerism , Structure-Activity Relationship , Urethra/drug effects , Urethra/physiology
10.
J Med Chem ; 42(23): 4794-803, 1999 Nov 18.
Article in English | MEDLINE | ID: mdl-10579842

ABSTRACT

Dihydropyrimidinones, such as 1, represent a novel class of alpha(1a) adrenoceptor antagonists with potential for the treatment of benign prostatic hyperplasia (BPH) (see part 1 of this series). Analysis of the metabolites of 1 revealed that 4-methoxycarbonyl-4-phenylpiperidine is formed as the major metabolite and is an agonist at the mu-opioid receptor. To circumvent any potential liability resulting from the metabolite, we decided to identify alternate templates devoid of agonist activity at the mu-opioid receptor to replace the 4-methoxycarbonyl-4-phenylpiperidine moiety. The present study describes the synthesis and SAR of dihydropyrimidinones linked to substituted 4-phenylpiperazine containing side chains. Compound (+)-38 was identified as a lead compound with a binding and functional profile comparable to that of 1. The putative metabolite 2-carboxamidophenylpiperazine has negligible affinity for the mu-opioid receptor.


Subject(s)
Adrenergic alpha-Antagonists/chemical synthesis , Piperazines/chemical synthesis , Pyrimidinones/chemical synthesis , Receptors, Adrenergic, alpha-1/metabolism , Adrenergic alpha-Antagonists/chemistry , Adrenergic alpha-Antagonists/metabolism , Adrenergic alpha-Antagonists/pharmacology , Animals , Binding, Competitive , Biological Availability , Dogs , Drug Design , GTP-Binding Proteins/metabolism , Half-Life , Humans , In Vitro Techniques , Male , Piperazines/chemistry , Piperazines/metabolism , Piperazines/pharmacology , Prostate/metabolism , Pyrimidinones/chemistry , Pyrimidinones/metabolism , Pyrimidinones/pharmacology , Rats , Receptors, Opioid, mu/agonists , Recombinant Proteins/metabolism , Stereoisomerism , Structure-Activity Relationship
11.
J Med Chem ; 42(23): 4804-13, 1999 Nov 18.
Article in English | MEDLINE | ID: mdl-10579843

ABSTRACT

We have previously disclosed dihydropyridines such as 1a,b as selective alpha(1a) antagonists as a potential treatment for benign prostatic hyperplasia (BPH). The propensity of dihydropyridines toward an oxidation led us to find suitable replacements of the core unit. The accompanying papers describe the structure-activity relationship (SAR) of dihydropyrimidinones 2a,b as selective alpha(1a) antagonists. We report herein the SAR of dihydropyrimidines such as 4 and highlight the similarities and differences between the dihydropyrimidine and dihydropyrimidinone series of compounds.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-Antagonists/chemical synthesis , Pyrimidines/chemical synthesis , Administration, Oral , Adrenergic alpha-Antagonists/chemistry , Adrenergic alpha-Antagonists/metabolism , Adrenergic alpha-Antagonists/pharmacology , Animals , Binding, Competitive , Biological Availability , Dogs , Drug Design , Drug Evaluation, Preclinical , Humans , In Vitro Techniques , Male , Prostate/metabolism , Pyrimidines/chemistry , Pyrimidines/metabolism , Pyrimidines/pharmacology , Rats , Receptors, Adrenergic, alpha-1/metabolism , Recombinant Proteins/metabolism , Stereoisomerism , Structure-Activity Relationship
12.
Drug Metab Dispos ; 27(11): 1360-6, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10534322

ABSTRACT

Tirofiban hydrochloride [L-tyrosine-N-(butylsulfonyl)-O-[4-(4-piperidinebutyl)] monohydrochloride, is a potent and specific fibrinogen receptor antagonist. Radiolabeled tirofiban was synthesized with either (3)H-label incorporated into the phenyl ring of the tyrosinyl residue or (14)C-label in the butane sulfonyl moiety. Neither human liver microsomes nor liver slices metabolized [(14)C]tirofiban. However, male rat liver microsomes converted a limited amount of the substrate to a more polar metabolite (I) and a relatively less polar metabolite (II). The formation of I was sex dependent and resulted from an O-dealkylation reaction catalyzed by CYP3A2. Metabolite II was identified as a 2-piperidone analog of tirofiban. There was no evidence for Phase II biotransformation of tirofiban by microsomes fortified with uridine-5'-diphospho-alpha-D-glucuronic acid. After a 1 mg/kg i.v. dose of [(14)C]tirofiban, recoveries of radioactivity in rat urine and bile were 23 and 73%, respectively. Metabolite I and unchanged tirofiban represented 70 and 30% of the urinary radioactivity, respectively. Tirofiban represented >90% of the biliary radioactivity. At least three minor biliary metabolites represented the remainder of the radioactivity. One of them was identified as I. Another was identified as II. When dogs received 1 mg/kg i.v. of [(3)H]tirofiban, most of the radioactivity was recovered in the feces as unchanged tirofiban. The plasma half-life of tirofiban was short in both rats and dogs, and tirofiban was not concentrated in tissues other than those of the vasculature and excretory organs.


Subject(s)
Fibrinolytic Agents/pharmacokinetics , Tyrosine/analogs & derivatives , Animals , Bile/metabolism , Dogs , Feces , Female , Fibrinolytic Agents/blood , Fibrinolytic Agents/urine , Half-Life , Humans , Male , Protein Binding , Rats , Rats, Sprague-Dawley , Tirofiban , Tissue Distribution , Tyrosine/blood , Tyrosine/pharmacokinetics , Tyrosine/urine
13.
J Clin Pharmacol ; 39(4): 385-93, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10197297

ABSTRACT

Alendronate is a potent bisphosphonate that has been studied for the treatment of osteoporosis and Paget's disease of the bone. To examine the pharmacokinetics of this drug, several groups of postmenopausal women were dosed intravenously in several studies. Twelve patients with metastatic bone disease were administered an intravenous dose of 10 mg of 14C-labeled alendronate (approximately 26 muCi), and plasma, feces, and urine samples were collected for 72 hours. Radioactivity was excreted almost exclusively in urine, and all of it was accounted for by alendronate. Overall recovery accounted for 47% of dose, with the remainder presumed to be retained in bone. Metabolism of alendronate was not observed. Renal clearance of alendronate was 71 mL/min. An additional 10 subjects were given repeated i.v. administrations of alendronate to demonstrate that previous exposure does not alter the pharmacokinetic behavior of the drug. Examination of the findings from these and other studies in which alendronate was administered intravenously revealed that disposition of single doses is linear in the range of 0.125 to 10 mg. With the possible exception of a somewhat greater skeletal retention of a systemically administered dose, the pharmacokinetics of i.v. alendronate were found to be similar to those of other bisphosphonates.


Subject(s)
Alendronate/pharmacokinetics , Adult , Aged , Alendronate/adverse effects , Alendronate/urine , Animals , Area Under Curve , Carbon Radioisotopes , Cricetinae , Dose-Response Relationship, Drug , Female , Fever/chemically induced , Headache/chemically induced , Humans , Infusions, Intravenous , Metabolic Clearance Rate , Middle Aged , Nausea/chemically induced , Postmenopause
14.
Drug Metab Dispos ; 26(5): 388-95, 1998 May.
Article in English | MEDLINE | ID: mdl-9571219

ABSTRACT

MK-499 [(+)-N-[1'-(6-cyano-1, 2, 3, 4-tetrahydro-2(R)-naphthalenyl)-3, 4-dihydro-4(R)-hydroxyspiro(2H-1-benzopyran-2, 4'-piperidin)-6-yl]methanesulfonamide] monohydrochloride is an investigational class III antiarrhythmic agent for treatment of malignant ventricular tachyarrhythmias. The disposition of [3H]MK-499 and [14C]MK-499 was studied in rats and dogs after oral and iv administration. MK-499 was concentrated in organs of excretion and the heart. In the rat, urinary radioactivity elimination values after iv (0.5 mg/kg) and oral (6.25 mg/kg) doses were 21 +/- 3% and 10 +/- 2%, respectively. Corresponding fecal recoveries were 68 +/- 6% and 78 +/- 7%. Similar results were found after corresponding doses of [14C]MK-499. In dogs, urine and feces accounted for 16 +/- 3% and 75 +/- 4% of recovered radioactivity after a [3H]MK-499 iv dose (0.1 mg/kg). Corresponding recoveries after an oral dose (1 mg/kg) were 12 +/- 2% and 76 +/- 3%. Biliary (0-24 hr) excretion accounted for 39 +/- 5% and 41 +/- 18% of [3H] and [14C] oral doses in rats, respectively. Dogs excreted 34% of [3H] oral dose in (0-24 hr) bile. The data indicated that a substantial amount of MK-499 was absorbed by rats and dogs. MK-499, metabolite I (formed by loss of N-substitution), and metabolite II (an acid formed by metabolic scission across the benzopyran ring) each represented 30% of rat urinary label. Rat bile contained MK-499 (10%), II (20%), and IV (10%), which was formed by carbon-4 hydroxylation of the tetralin ring. Additionally, rat bile included glutathione (V) and N-acetyl-1-cysteine (VI) conjugates of a ring-opened metabolite. Metabolite III, a positional isomer of IV, was excreted in rat urine. The major labeled species excreted in dog bile were unchanged MK-499 and its glucuronide (VII), which, respectively, represented 50% and 30% of the biliary radioactivity. MK-499 and a small amount of I represented dog urinary radioactivity. The bioavailability of MK-499 was high in dogs (100%) but low in rats (17%). This difference was probably due to the more extensive presystemic metabolism of MK-499 in rats.


Subject(s)
Anti-Arrhythmia Agents/pharmacokinetics , Benzopyrans/pharmacokinetics , Piperidines/pharmacokinetics , Tachycardia, Ventricular/metabolism , Animals , Anti-Arrhythmia Agents/therapeutic use , Benzopyrans/therapeutic use , Biological Availability , Chromatography, High Pressure Liquid , Dogs , Dose-Response Relationship, Drug , Magnetic Resonance Spectroscopy , Male , Metabolic Clearance Rate , Piperidines/therapeutic use , Rats , Rats, Sprague-Dawley , Tachycardia, Ventricular/drug therapy , Tissue Distribution
15.
Drug Metab Dispos ; 25(10): 1191-9, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9321523

ABSTRACT

Simvastatin (SV) is a lactone prodrug used for the treatment of hypercholesterolemia. Upon incubation of SV with liver microsomal preparations from human donors, four major metabolic products were formed (3'-hydroxy SV, 6'-exomethylene SV, 3',5'-dihydrodiol SV, and the active hydroxy acid, SVA), together with several minor unidentified metabolites. The 3',5'-dihydrodiol SV, a new metabolite, was inactive as an inhibitor of HMG-CoA reductase. Kinetic studies of SV metabolism in human liver microsomes suggested that the major NADPH-dependent metabolites (3'-hydroxy SV, 6'-exomethylene SV, and 3',5'-dihydrodiol SV) were formed with relatively high intrinsic clearances, consistent with the extensive metabolism of SV observed in vivo. Based on four different in vitro approaches, namely 1) correlation analysis, 2) chemical inhibition, 3) immunoinhibition, and 4) metabolism by recombinant human P450, it is concluded that CYP3A is the major enzyme subfamily responsible for the metabolism of SV by human liver microsomes. Both CYP3A4 and CYP3A5 were capable of catalyzing the formation of 3',5'-dihydrodiol, 3'-hydroxy, and 6'-exomethylene metabolites. However, CYP3A4 exhibited higher affinity (> 3 fold) for SV than CYP3A5. Also, the studies indicated that CYP2D6, CYP2A6, CYP2C8, CYP2C9, CYP2C19, CYP1A2, and CYP2E1 did not play significant roles in the metabolism of SV in vitro. Over the concentration range of 0-40 microM, SV inhibited the activity of CYP3A, but not the activities of CYP2C8/9, CYP2C19, or CYP2D6 in human liver microsomes. The inhibition of hepatic midazolam 1'-hydroxylase, a CYP3A marker activity, by SV was competitive with a Ki value of approximately 10 microM. SV was > 30-fold less potent than ketoconazole and itraconazole as an inhibitor of CYP3A. Under the same conditions, SVA, the hydrophilic hydroxy acid form of SV, did not inhibit CYP3A, CYP2C8/9, CYP2C19, or CYP2D6 activities. The results suggested that the in vivo inhibitory effects of SV on the metabolism of CYP3A substrates likely would be less than those of ketoconazole and itraconazole at their respective therapeutic concentrations. In addition, metabolic activities mediated by the other P450 enzymes tested are unlikely to be affected by SV.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/metabolism , Simvastatin/metabolism , Anticholesteremic Agents/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Microsomes, Liver/drug effects , Simvastatin/pharmacology
16.
Drug Metab Dispos ; 25(8): 978-84, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9280406

ABSTRACT

The present study demonstrates the utility of an in vitro-in vivo correlative approach in the selection of an optimum prodrug candidate of L-767,679 (N-([7-(piperazin-1-yl)-3,4-dihydro-1(1H)-isoquinolinone-2-yl]acetyl)-3(S)-(ethynyl)-beta-alanine), a potent fibrinogen receptor antagonist. As an initial screening step, a comparative in vitro hepatic metabolism study was conducted for L-767,679 and a series of aliphatic and aromatic ester prodrugs in dogs, monkeys, and humans. In all species, the active acid L-767,679, but not the ester prodrugs, was resistant to metabolism. Only the methyl, ethyl, and isopropyl esters were converted exclusively to the active acid in liver microsomal preparations from dogs and humans, and thus were selected for further studies. In the preparations from monkeys, all of the esters investigated were metabolized efficiently to both the active acid and several other products. The absolute formation rates of L-767,679 from the esters followed the rank order: methyl approximately ethyl > isopropyl in all species, and in humans > dogs for the three esters. The three ester prodrugs did not undergo appreciable hydrolysis in blood or upon incubation with intestinal S9 from any of the studied species. In vivo evaluation of the previous three aliphatic esters in dogs and monkeys supported the in vitro findings. L-767,679 was metabolically stable in both dogs and monkeys. After intravenous administration of the prodrugs to either species, the extent of acid formation was higher in dogs than in monkeys. In addition, the extent of L-767,679 formed from these prodrugs followed the rank order: methyl approximately ethyl > isopropyl. Similar results were obtained after oral dosing of the prodrugs, such that the bioavailability of L-767,679 was higher in dogs than in monkeys, and the bioavailability was higher after the ethyl ester than after the isopropyl prodrug in both species. In either species, both ethyl and isopropyl ester prodrugs were better absorbed than L-767,679. Overall, the results suggested that the bioavailability of the active acid after administration of an ester prodrug was dictated primarily by two factors, viz.:1) the relative rates of ester hydrolysis versus competing metabolic reactions and 2) the absolute rates of ester hydrolysis. In the case of L-767,679 prodrugs, absorption was not a limiting factor. Consequently, the bioavailability of L-767,679 after oral administration of the ester prodrugs would likely be greater in humans than in dogs, and in humans would be higher with the ethyl ester than with the isopropyl ester. On this basis, the ethyl ester was considered as a promising candidate for clinical evaluation as a fibrinogen receptor antagonist prodrug.


Subject(s)
Piperazines/pharmacokinetics , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Prodrugs/pharmacokinetics , beta-Alanine/analogs & derivatives , Administration, Oral , Animals , Biological Availability , Chromatography, High Pressure Liquid , Dogs , Humans , Macaca mulatta , Male , Microsomes, Liver/metabolism , Piperazines/metabolism , Prodrugs/metabolism , Species Specificity , beta-Alanine/metabolism , beta-Alanine/pharmacokinetics
17.
Drug Metab Dispos ; 25(3): 355-61, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9172954

ABSTRACT

L-738,167 is a potent and long-acting fibrinogen receptor antagonist and may be useful for treatment of chronic thrombotic occlusive disorders. The purposes of this study were to characterize the metabolism and disposition of L-738,167, and to investigate factors affecting its pharmacokinetic behaviors in dogs, one of the animal models used in pharmacological and toxicological studies. In vitro and in vivo experiments indicated that L-738,167 was not metabolized to any appreciable extent in dogs. Biliary excretion was found to be the major route (approximately 75%) of drug elimination. Following 1 and 3 micrograms/kg iv doses, blood pharmacokinetics of L-738,167 were linear. Total blood clearance (CLB) was much lower than hepatic blood flow, and the apparent volume of distribution at steady-state (Vdss,B) was comparable with blood volume. Blood pharmacokinetics in the dose range of 3-250 micrograms/kg were dose-dependent; both CLB and Vdss,B for L-738,167 increased markedly with increasing doses. However, the terminal half-life (t1/2) was dose-independent, with a mean value of approximately 4 days. L-738,167 was found to bind negligibly to dog plasma proteins. Determinations of whole blood (WB), platelet-rich plasma, and platelet-poor plasma concentrations after several intravenous doses of [3H]L-738,167 revealed significant concentration-dependent binding of the compound to platelets. Kinetic analysis of the platelet binding indicated that L-738,167 was bound to dog platelets with high affinity (apparent Kd approximately 1 nM platelet-poor plasma concentration) and relatively low capacity (approximately 70 nM WB concentration). Findings are consistent with the binding kinetics of L-738,167 to glycoprotein IIb/IIIa (GP IIb/IIIa) receptor, supporting that GP IIb/IIIa was the primary binding component on the platelets. It was concluded that the dose-dependent pharmacokinetics of L-738,167 were the consequence of the concentration-dependent drug-platelet binding. Due to this extensive platelet binding, L-738,167, when given in therapeutic doses or lower, resided primarily in the vascular compartment-the site of pharmacological action. At doses exceeding the receptor binding capacity, the excess amount or the unbound drug was eliminated rapidly. In all cases, the equally long t1/2 of L-738,167 was also a consequence of the high-affinity binding to platelets, in good agreement with its prolonged pharmacodynamic profile.


Subject(s)
Azepines/pharmacokinetics , Fibrinolytic Agents/pharmacokinetics , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Sulfonamides/pharmacokinetics , Animals , Azepines/blood , Azepines/pharmacology , Blood Platelets/metabolism , Blood Proteins/metabolism , Dogs , Dose-Response Relationship, Drug , Fibrinolytic Agents/blood , Fibrinolytic Agents/pharmacology , Injections, Intravenous , Male , Protein Binding , Sulfonamides/blood , Sulfonamides/pharmacology , Tritium
18.
Drug Metab Dispos ; 24(11): 1263-71, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8937862

ABSTRACT

The pharmacokinetics and bioavailability of L-751,164, an ethyl ester prodrug of a potent fibrinogen receptor antagonist, L-742,998, were studied in beagle dogs and rhesus monkeys. In both species, L-751,164 exhibited high clearance. After an intravenous dose, L-751,164 was converted to the parent L-742,998 to the extent of approximately 20% in dogs and 90% in monkeys. After oral administration of the prodrug, however, the bioavailability, measured either as the prodrug or as the active parent, was < 5% in both species. Several experiments were conducted subsequently to investigate possible causes for the observed similarities in the low oral bioavailability of the prodrug between species despite its differences in the in vivo conversion. In vitro metabolism studies using dog liver subcellular fractions indicated extensive metabolism of L-751,164 to metabolites other than L-742,998. Kinetically, L-742,998 formation accounted only for approximately 25% of the prodrug disappearance. In contrast, monkey liver preparations converted L-751,164 exclusively and rapidly to L-742,998. Good agreement between the in vitro hepatic metabolism and the in vivo observations suggests that liver was the major eliminating organ after intravenous administration of the prodrug in both species. In dogs, this suggestion was further supported by low bioavailability of the prodrug (20%) and the parent (below detection limit) after intraportal administration of the prodrug. In vitro metabolism of L-751,164 using intestinal S9 fractions revealed substantial metabolism in monkeys, but not in dogs. Several NADPH-dependent metabolites were observed with monkey intestinal preparation, with the parent L-742,998 being the minor product (approximately 25-30%). Furthermore, L-751,164 was shown, by means of an in vitro Caco-2 cell, and in situ rat intestinal loop models, to be highly permeable to intestinal barriers. Collectively, these results suggest that the apparent species differences in the prodrug conversion observed in vivo likely were due to species differences in the hepatic metabolism of the prodrug. In both species, the high first-pass metabolism of the prodrug, and the extensive conversion of the prodrug to metabolic products other than the parent contributed, at least in part, to the low bioavailability of the prodrug and active parent, respectively, obtained after an oral dose of the prodrug. The latter process was species-dependent, involving primarily the hepatic first-pass elimination in dogs and the intestinal first-pass metabolism in monkeys.


Subject(s)
Prodrugs/pharmacokinetics , Pyridines/pharmacokinetics , Animals , Biological Availability , Caco-2 Cells , Dogs , Humans , In Vitro Techniques , Intestinal Mucosa/metabolism , Liver/metabolism , Macaca mulatta , Male , Species Specificity , Tissue Distribution
19.
Drug Metab Dispos ; 24(6): 634-42, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8781778

ABSTRACT

Drug-metabolizing enzymes were studied in subcellular fractions of dog, monkey, and human small intestines, and in the human adenocarcinoma cell line Caco-2, a commonly used in vitro absorption model. Immunoblot analysis indicated the presence of enzymes related to cytochrome P450 (CYP) 1A1/CYP1A2, CYP2D6, CYP3A, and carboxylesterases (ESs) in human and monkey intestines, and of CYP3A and ES in dog intestines. Catalytically, human and monkey intestines exhibited significant and comparable testosterone 6 beta-hydroxylase, (+)-bufuralol 1'-hydroxylase, and ES activities. In contrast, dog intestine possessed moderate testosterone 6 beta-hydroxylase, much lower ES, and undetectable bufuralol hydroxylase activities. In addition, low tolbutamide methylhydroxylase activity was observed in human and monkey intestines, but not in dog intestines. Of the phase I enzymes investigated, only ES was detected immunologically and functionally in Caco-2 cells. With respect to phase II enzymes, human and monkey intestines contained relatively high intestinal glucuronyltransferase, N-acetyltransferase (NAT), sulfotransferase, and glutathione S-transferase activities. Except for NAT, all phase II enzymes studied were detectable in dog intestines. In Caco-2 cells, acetaminophen sulfation activity was below the limit of detection, whereas all other conjugating activities were evident. Studies of enzyme kinetics and inhibition by known inhibitors of testosterone 6 beta-hydroxylase activity, the major intestinal mono-oxygenase in all species, revealed some similarities between the responsible enzymes. Comparative studies with human liver microsomes suggested the possible involvement of CYP3A enzymes in the intestinal catalysis of testosterone 6 beta-hydroxylation similar to those observed with human hepatic CYP3A. Further studies on ESs, however, revealed multiplicity and species and/or tissue differences in the microsomal and cytosolic enzymes. Based on kinetic studies, monkey intestines and Caco-2 cells possessed NAT activities, with properties similar to those in human intestine and liver. Overall, the results demonstrated that both the preparations of small intestines and Caco-2 cells exhibited significant drug-metabolizing enzyme activities, although several differences were noted between the intestinal enzymes in the animals or in the Caco-2 cells and those found in humans.


Subject(s)
Cytochrome P-450 CYP1A2/analysis , Cytochrome P-450 CYP2D6/analysis , Cytochrome P-450 Enzyme System/analysis , Intestine, Small/enzymology , Steroid Hydroxylases/analysis , Adult , Aged , Animals , Caco-2 Cells/drug effects , Caco-2 Cells/enzymology , Dogs , Humans , In Vitro Techniques , Macaca mulatta , Male , Microsomes, Liver/enzymology , Middle Aged , Species Specificity
20.
Chem Res Toxicol ; 9(2): 547-54, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8839061

ABSTRACT

The novel 5-lipoxygenase inhibitor [1S,5R]-3-cyano-1-(3-furyl)-6-(6-[3-(3 alpha-hydroxy-6,8-dioxabicyclo[3.2.1]octanyl)]pyridin-2-yl- methoxyl)naphthalene (L-739,010), when administered to rats and rhesus monkeys, was found to produce metabolites which appeared to be covalently bound to plasma proteins. Incubation of [14C]L-739,010 with rat liver microsomes did not yield appreciable amounts of soluble metabolites but resulted in covalent binding to microsomal proteins. The covalent binding was NADPH-dependent and was enhanced by 1.5- and 2-fold in liver microsomes from rats, pretreated with phenobarbital and dexamethasone, respectively. Addition of triacetyloleandomycin and diethyldithiocarbamate to the incubation mixture inhibited the covalent binding by 60% and 46%, respectively. These findings suggest that the cytochrome P450 3A family of enzymes play an important role in the bioactivation of L-739,010. The presence of GSH attenuated the covalent binding by 50%, while methoxylamine, an aldehyde trapping agent, blocked the covalent binding completely and, concurrently, produced several soluble metabolic adducts. Subsequently, major methoxylamine adducts were identified by LC-MS/MS and NMR as O-methyloximes of the ring-opened furan moiety of L-739,010. Incubation of L-739,010 with methoxylamine and hepatic microsomes from dog, rhesus monkey, and human produced similar metabolic adducts as those formed by rat liver microsomes. Therefore, under these experimental conditions, the furan moiety, which undergoes oxidative cleavage to the highly reactive 2-butene-1,4-dialdehyde, represents the major site of L-739,010 biotransformation. This putative reactive intermediate could react with microsomal proteins in vitro and physiological proteins in vivo. Since furan bioactivation is believed to be responsible for the toxicity of many furan-containing compounds, the furan moiety of L-739,010 may be regarded as undesirable.


Subject(s)
Bridged Bicyclo Compounds/pharmacokinetics , Furans/pharmacokinetics , Lipoxygenase Inhibitors/pharmacokinetics , Microsomes, Liver/metabolism , Quinolines/pharmacokinetics , Animals , Biotransformation , Dogs , Humans , Macaca mulatta , Mass Spectrometry , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...