Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Pharmacol ; 59(1): 139-152, 2019 01.
Article in English | MEDLINE | ID: mdl-30192390

ABSTRACT

Maraviroc is a C-C chemokine receptor type-5 antagonist approved for the treatment of HIV-1. Previous studies show that cytochrome P450 3A5 (CYP3A5) plays a role in maraviroc metabolism. CYP3A5 is subject to a genetic polymorphism. The presence of 2 functional alleles (CYP3A5*1/*1) confers the extensive metabolism phenotype, which is rare in whites but common in blacks. The effect of CYP3A5 genotype on maraviroc and/or metabolite pharmacokinetics was evaluated in 2 clinical studies: a post hoc analysis from a phase 2b/3 study (NCT00098293) conducted in 494 HIV-1-infected subjects (study 1) in which the impact on maraviroc efficacy in 303 subjects was also assessed, and a study conducted in 47 healthy volunteers (study 2). In study 2 (NCT02625207), extensive metabolizers had 26% to 37% lower mean area under the concentration-time curve compared with poor metabolizers (no CYP3A5*1 alleles). This effect diminished to 17% in the presence of potent CYP3A inhibition. The effect of CYP3A5 genotype was greatest in the formation of the metabolite (1S,2S)-2-hydroxymaraviroc. In study 1, the CYP3A5*1/*1 genotype unexpectedly had higher maraviroc area under the curve predictions (20%) compared with those with no CYP3A5*1 alleles. The reason for this disparity remains unclear. The proportions of subjects with viral loads <50 and <400 copies/mL for maraviroc were comparable among all 3 CYP3A5 genotypes. In both studies maraviroc exposures were in the range of near-maximal viral inhibition in the majority of subjects. These results demonstrate that although CYP3A5 contributes to the metabolism of maraviroc, CYP3A5 genotype does not affect the clinical response to maraviroc in combination treatment of HIV-1 infection at approved doses.


Subject(s)
Cytochrome P-450 CYP3A/genetics , HIV Fusion Inhibitors/pharmacokinetics , HIV Fusion Inhibitors/therapeutic use , HIV Infections , HIV-1 , Maraviroc/pharmacokinetics , Maraviroc/therapeutic use , Adult , Double-Blind Method , Female , Genotype , HIV Fusion Inhibitors/blood , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/metabolism , Healthy Volunteers , Humans , Male , Maraviroc/blood , Middle Aged , Polymorphism, Genetic , Treatment Outcome , Young Adult
2.
Drug Metab Dispos ; 46(5): 493-502, 2018 May.
Article in English | MEDLINE | ID: mdl-29475834

ABSTRACT

Maraviroc (MVC) is a CCR5 coreceptor antagonist indicated in combination with other antiretroviral agents for the treatment of CCR5-tropic human immunodefinciency virus-1 infection. In this study, the metabolism of MVC was investigated in human liver microsomes to delineate the relative roles of CYP3A4 and CYP3A5. MVC is metabolized to five hydroxylated metabolites, all of which were biosynthesized and identified using mass and NMR spectroscopy. The sites of metabolism were the 2- and 3-positions of the 4,4-difluorocyclohexyl moiety and the methyl of the triazole moiety. Absolute configurations were ultimately ascertained by comparison to authentic standards. The biosynthesized metabolites were used for quantitative in vitro experiments in liver microsomes using cyp3cide, a selective inactivator of CYP3A4. (1S,2S)-2-OH-MVC was the main metabolite representing approximately half of the total metabolism, and CYP3A5 contributed approximately 40% to that pathway in microsomes from CYP3A5*1/*1 donors. The other four metabolites were almost exclusively metabolized by CYP3A4. (1S,2S)-2-hydroxylation also correlated to T-5 N-oxidation, a CYP3A5-specific activity. These data are consistent with clinical pharmacokinetic data wherein CYP3A5 extensive metabolizer subjects showed a modestly lower exposure to MVC.


Subject(s)
Cyclohexanes/metabolism , Cytochrome P-450 CYP3A/metabolism , Triazoles/metabolism , Cyclohexanes/pharmacokinetics , Humans , Hydroxylation/physiology , Kinetics , Maraviroc , Microsomes, Liver/metabolism , Oxidation-Reduction , Pyrazoles/metabolism , Pyrazoles/pharmacokinetics , Pyrimidines/metabolism , Pyrimidines/pharmacokinetics , Triazoles/pharmacokinetics
3.
Drug Metab Lett ; 4(3): 162-72, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20642449

ABSTRACT

The quinuclidine PHA-0568487(1) is an agonist of the alpha 7 nicotinic acetylcholine receptor that was designed to mitigate the bioactivation associated with the core scaffold and subsequently remove associated liabilities with in vivo tolerability. The drug metabolites of 1 in nonclinical species were identified in plasma and urine of rats, dogs and monkeys receiving oral administrations of 1. The in vitro biotransformation of 1 was subsequently investigated in multiple species employing cryopreserved hepatocytes, hepatic subcellular fractions and recombinantly-expressed human P450 enzymes. In addition, in vitro metabolism of synthetically prepared metabolite precursors were instrumental in the elucidation of several secondary metabolites. The results indicated that the principal biotransformation of 1 was oxidation of the benzo[1,4]dioxane moiety (M8, M10) followed by subsequent oxidation to a range of secondary metabolites (M1-7, M9, M11, M13-15, and M17-18). The carboxylic acids M1 and M2 resulting from the oxidative cleavage of the dioxane ring were the principal metabolites observed in the plasma, urine and hepatocyte incubations across all species (M1 & M2). Quinuclidine oxidation was another pathway of importance, yielding an N-oxide (M12) which was also observed in all species.P450 2D6 and FMO1 catalyze the oxidation of the quinuclidine nitrogen. The N oxidation of the quinuclidine moiety is consistent with previously published accounts of this scaffold's metabolism and, interestingly, may implicate the uncommon quinuclidine moiety as an entity directing the metabolism of this scaffold (e.g., 1) via FMO1 and P450 2D6 oxidation.


Subject(s)
Aza Compounds/pharmacokinetics , Dioxins/pharmacokinetics , Nicotinic Agonists/pharmacokinetics , Quinuclidines/pharmacokinetics , Receptors, Nicotinic/drug effects , Administration, Oral , Animals , Aza Compounds/administration & dosage , Aza Compounds/blood , Aza Compounds/urine , Biotransformation , Chromatography, Liquid , Cytochrome P-450 CYP2D6/metabolism , Dioxins/administration & dosage , Dioxins/blood , Dioxins/urine , Dogs , Haplorhini , Hepatocytes/enzymology , Humans , Magnetic Resonance Spectroscopy , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/blood , Nicotinic Agonists/urine , Oxidation-Reduction , Oxygenases/metabolism , Quinuclidines/administration & dosage , Quinuclidines/blood , Quinuclidines/urine , Rats , Recombinant Proteins/metabolism , Tandem Mass Spectrometry , alpha7 Nicotinic Acetylcholine Receptor
4.
ACS Med Chem Lett ; 1(5): 219-23, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-24900198

ABSTRACT

As part of a strategy to deliver short-acting calcium-sensing receptor (CaSR) antagonists, the metabolically labile thiomethyl functionality was incorporated into the zwitterionic amino alcohol derivative 3 with the hope of increasing human clearance through oxidative metabolism, while delivering a pharmacologically inactive sulfoxide metabolite. The effort led to the identification of thioanisoles 22 and 23 as potent and orally active CaSR antagonists with a rapid onset of action and short pharmacokinetic half-lives, which led to a rapid and transient stimulation of parathyroid hormone in a dose-dependent fashion following oral administration to rats. On the basis of the balance between target pharmacology, safety, and human disposition profiles, 22 and 23 were advanced as clinical candidates for the treatment of osteoporosis.

5.
Drug Metab Dispos ; 34(9): 1615-23, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16790552

ABSTRACT

The metabolism and excretion of N-(3R)-1-azabicyclo[2.2.2]oct-3-ylfuro[2,3-c]pyridine-5-carboxamide (1), an agonist of the alpha7 nicotinic acetylcholinergic receptor, were determined in both Sprague-Dawley rats and beagle dogs using [3H]1. Initially, 3-tritio-furanopyridine 1 ([3H]1a) was evaluated in pilot mass balance studies by determining total radioactivity recovery and pharmacokinetics in lyophilized excreta and nonlyophilized plasma, respectively. Lower mass balance and much greater circulatory radioactivity exposures were observed in rats than in dogs, with urinary tritiated water (HTO) only detected in rats. The 133-h half-life in rats, possibly due to very slowly eliminated metabolites, was more likely attributable to HTO formed from [3H]1a because of site-specific chemical and/or metabolic 3H instability, which was confirmed by urinary HTO. In contrast, dog data supported 3H stability within [3H]1a. Conflicting cross-species data with [3H]1a suggested species-specific metabolic fates for 1, requiring a 3H form of 1 resistant to 3H loss in rats. Therefore, tritiation of 1 at its furanopyridine C7, a site predicted to be both chemically and metabolically stable, yielded 7-tritio-N-(3R)-1-azabicyclo[2.2.2]oct-3-ylfuro[2,3-c]pyridine-5-carboxamide ditrifluoroacetate ([3H]1b), which allowed in both species the determination of all excretory pathways, total radioactivity pharmacokinetics, and major excretory and circulatory metabolites with complete radioactivity recovery without HTO generation. Definitive metabolite elucidation for 1 using [3H]1b confirmed the suspected species-dependent metabolic susceptibility for 3H loss from [3H]1a in rats, but not dogs, since the majority of rat metabolites resulted from furanopyridine biotransformation. The described studies explore the evaluation of tritium exchange risk from a mechanistic biotransformation perspective and highlight the need for careful deliberation when considering and designing 3H compounds for radiolabeled metabolism studies.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Drug Evaluation, Preclinical , Isotope Labeling , Quinuclidines/pharmacokinetics , Tritium , Animals , Biotransformation , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Dogs , Drug Evaluation, Preclinical/methods , Drug Stability , Feces/chemistry , Molecular Structure , Quinuclidines/chemistry , Rats , Rats, Sprague-Dawley , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...