Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
Br J Clin Pharmacol ; 84(1): 130-141, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28850715

ABSTRACT

AIMS: BMS-823778 is an inhibitor of 11ß-hydroxysteroid dehydrogenase type-1, and thus a potential candidate for Type 2 diabetes treatment. Here, we investigated the metabolism and pharmacokinetics of BMS-823778 to understand its pharmacokinetic variations in early clinical trials. METHODS: The metabolism of BMS-823778 was characterized in multiple in vitro assays. Pharmacokinetics were evaluated in healthy volunteers, prescreened as CYP2C19 extensive metabolizers (EM) or poor metabolizers (PM), with a single oral dose of [14 C]BMS-823778 (10 mg, 80 µCi). RESULTS: Three metabolites (<5%) were identified in human hepatocytes and liver microsomes (HLM) incubations, including two hydroxylated metabolites (M1 and M2) and one glucuronide conjugate (M3). As the most abundant metabolite, M1 was formed mainly through CYP2C19. M1 formation was also correlated with CYP2C19 activities in genotyped HLM. In humans, urinary excretion of dosed radioactivity was significantly higher in EM (68.8%; 95% confidence interval 61.3%, 76.3%) than in PM (47.0%; 43.5%, 50.6%); only small portions (<2%) were present in faeces or bile from both genotypes. In plasma, BMS-823778 exposure in PM was significantly (5.3-fold, P = 0.0097) higher than in EM. Furthermore, total radioactivity exposure was significantly higher (P < 0.01) than BMS-823778 exposure in all groups, indicating the presence of metabolites. M1 was the only metabolite observed in plasma, and much lower in PM. In urine, the amount of M1 and its oxidative metabolite in EM was 7-fold of that in PM, while more glucuronide conjugates of BMS-823778 and M1 were excreted in PM. CONCLUSIONS: CYP2C19 polymorphisms significantly impacted systemic exposure and metabolism pathways of BMS-823778 in humans.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Cytochrome P-450 CYP2C19/genetics , Polymorphism, Genetic , Pyridines/pharmacokinetics , Renal Elimination/genetics , Triazoles/pharmacokinetics , Adult , Drug Administration Schedule , Genotype , Glucuronides/metabolism , Healthy Volunteers , Hepatocytes/metabolism , Humans , Hydroxylation , Male , Microsomes, Liver/metabolism , Oxidation-Reduction , Pyridines/administration & dosage , Pyridines/blood , Pyridines/metabolism , Triazoles/administration & dosage , Triazoles/blood , Triazoles/metabolism , Young Adult
2.
Br J Clin Pharmacol ; 76(3): 432-44, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23210765

ABSTRACT

AIM(S): This study assessed the effect of differences in renal function on the pharmacokinetics and pharmacodynamics of dapagliflozin, a renal sodium glucose co-transporter-2 (SGLT2) inhibitor for the treatment of type 2 diabetes mellitus (T2DM). METHODS: A single 50 mg dose of dapagliflozin was used to assess pharmacokinetics and pharmacodynamics in five groups: healthy non-diabetic subjects; patients with T2DM and normal kidney function and patients with T2DM and mild, moderate or severe renal impairment based on estimated creatinine clearance. Subsequently, 20 mg once daily multiple doses of dapagliflozin were evaluated in the patients with T2DM. Formation rates of dapagliflozin 3-O-glucuronide (D3OG), an inactive metabolite, were evaluated using human isolated kidney and liver microsomes. RESULTS: Plasma concentrations of dapagliflozin and D3OG were incrementally increased with declining kidney function. Steady-state Cmax for dapagliflozin were 4%, 6% and 9% higher and for D3OG were 20%, 37% and 52% higher in patients with mild, moderate and severe renal impairment, respectively, compared with normal function. AUC(0,τ) was likewise higher. D3OG formation in kidney microsomes was three-fold higher than in liver microsomes and 109-fold higher than in intestine microsomes. Compared with patients with normal renal function, pharmacodynamic effects were attenuated with renal impairment. Steady-state renal glucose clearance was reduced by 42%, 83% and 84% in patients with mild, moderate or severe renal impairment, respectively. CONCLUSIONS: These results indicate that both kidney and liver significantly contribute to dapagliflozin metabolism, resulting in higher systemic exposure with declining kidney function. Dapagliflozin pharmacodynamics in diabetic subjects with moderate to severe renal impairment are consistent with the observation of reduced efficacy in this patient population.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucosides/therapeutic use , Hypoglycemic Agents/therapeutic use , Kidney/physiopathology , Sodium-Glucose Transporter 2 Inhibitors , Adolescent , Adult , Aged , Benzhydryl Compounds , Blood Glucose/analysis , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/urine , Dose-Response Relationship, Drug , Female , Glucosides/adverse effects , Glucosides/pharmacokinetics , Glucosides/pharmacology , Glycosuria/urine , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Kidney/drug effects , Kidney/metabolism , Kidney Function Tests , Liver/drug effects , Liver/metabolism , Liver/physiopathology , Male , Middle Aged , Sodium-Glucose Transporter 2 , Young Adult
3.
Br J Clin Pharmacol ; 70(6): 870-80, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21175442

ABSTRACT

AIM: To further explore the mechanism underlying the interaction between repaglinide and gemfibrozil, alone or in combination with itraconazole. METHODS: Repaglinide metabolism was assessed in vitro (human liver subcellular fractions, fresh human hepatocytes, and recombinant enzymes) and the resulting incubates were analyzed, by liquid chromatography-mass spectrometry (LC-MS) and radioactivity counting, to identify and quantify the different metabolites therein. Chemical inhibitors, in addition to a trapping agent, were also employed to elucidate the importance of each metabolic pathway. Finally, a panel of human liver microsomes (genotyped for UGT1A1*28 allele status) was used to determine the importance of UGT1A1 in the direct glucuronidation of repaglinide. RESULTS: The results of the present study demonstrate that repaglinide can undergo direct glucuronidation, a pathway that can possibly contribute to the interaction with gemfibrozil. For example, [³H]-repaglinide formed glucuronide and oxidative metabolites (M2 and M4) when incubated with primary human hepatocytes. Gemfibrozil effectively inhibited (∼78%) both glucuronide and M4 formation, but had a minor effect on M2 formation. Concomitantly, the overall turnover of repaglinide was also inhibited (∼80%), and was completely abolished when gemfibrozil was co-incubated with itraconazole. These observations are in qualitative agreement with the in vivo findings. UGT1A1 plays a significant role in the glucuronidation of repaglinide. In addition, gemfibrozil and its glucuronide inhibit repaglinide glucuronidation and the inhibition by gemfibrozil glucuronide is time-dependent. CONCLUSIONS: Inhibition of UGT enzymes, especially UGT1A1, by gemfibrozil and its glucuronide is an additional mechanism to consider when rationalizing the interaction between repaglinide and gemfibrozil.


Subject(s)
Carbamates/pharmacology , Gemfibrozil/pharmacology , Hypoglycemic Agents/pharmacology , Hypolipidemic Agents/pharmacology , Piperidines/pharmacology , Biotransformation , Carbamates/pharmacokinetics , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Glucuronides/metabolism , Glucuronosyltransferase/physiology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hypoglycemic Agents/pharmacokinetics , Itraconazole/pharmacology , Microsomes, Liver/metabolism , Piperidines/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL