Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
ACS Med Chem Lett ; 11(12): 2504-2509, 2020 Dec 10.
Article in English | MEDLINE | ID: mdl-33335674

ABSTRACT

The 6-benzhydryl-4-amino-quinolin-2-ones are peripherally restricted CB1 receptor inverse agonists (CB1RIAs) that have been reported to attenuate obesity and improve insulin sensitivity in the diet-induced obese (DIO) mouse model. However, chronic dosing of select compounds from the series showed time-dependent brain accumulation despite a low brain/plasma exposure ratio. To address this issue, a PEGylation approach was employed to identify a novel series of homodimeric 6-benzhydryl-4-amino-quinazoline-PEG conjugates with an extended half-life. The lead compound 18 engaged peripheral CB1Rs in a gastrointestinal (GI) tract motility study and demonstrated a high level of peripheral restriction in a chronic DIO mouse pharmacokinetic study.

2.
Bioorg Med Chem Lett ; 30(17): 127387, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32738984

ABSTRACT

(2S,3R,4R,5S,6R)-2-Aryl-5,5-difluoro-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4-diols and (2S,3R,4R,5S,6R)-2-aryl-5-fluoro-5-methyl-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4-diols were discovered as dual inhibitors of sodium glucose co-transporter proteins (e.g. SGLT1 and SGLT2) through rational drug design, efficient synthesis, and in vitro and in vivo evaluation. Compound 6g demonstrated potent dual inhibitory activities (IC50 = 96 nM for SGLT1 and IC50 = 1.3 nM for SGLT2). It showed robust inhibition of blood glucose excursion in an oral glucose tolerance test (OGTT) in Sprague Dawley (SD) rats when dosed at both 1 mg/kg and 10 mg/kg orally. It also demonstrated postprandial glucose control in db/db mice when dosed orally at 10 mg/kg.


Subject(s)
Glucosides/chemistry , Hypoglycemic Agents/chemistry , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2/chemistry , Sodium-Glucose Transporter 2/metabolism , Animals , Blood Glucose/analysis , Diabetes Mellitus/drug therapy , Diabetes Mellitus/pathology , Disease Models, Animal , Drug Design , Glucose Tolerance Test , Glucosides/metabolism , Glucosides/pharmacology , Glucosides/therapeutic use , Half-Life , Halogenation , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Inhibitory Concentration 50 , Mice , Mice, Inbred C57BL , Microsomes/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 1/metabolism , Structure-Activity Relationship
3.
Bioorg Med Chem Lett ; 29(20): 126668, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31519374

ABSTRACT

Type 2 diabetes mellitus (T2DM) is characterized by chronically elevated plasma glucose levels. The inhibition of glucagon-induced hepatic glucose output via antagonism of the glucagon receptor (GCGR) using a small-molecule antagonist is a promising mechanism for improving glycemic control in the diabetic state. The present work discloses the discovery of indazole-based ß-alanine derivatives as potent GCGR antagonists through an efficient enantioselective synthesis and structure-activity relationship (SAR) exploration and optimization. Compounds within this class exhibited excellent pharmacokinetic properties in multiple preclinical species. In an acute dog glucagon challenge test, compound 13K significantly inhibited glucagon-mediated blood glucose increase when dosed orally at 10 mg/kg.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/chemical synthesis , Indazoles/chemistry , Receptors, Glucagon/antagonists & inhibitors , beta-Alanine/chemical synthesis , Amino Acid Sequence , Animals , Blood Glucose/drug effects , Carbohydrate Metabolism , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Hypoglycemic Agents/pharmacokinetics , Liver/metabolism , Mice , Models, Molecular , Molecular Structure , Protein Binding , Rats , Structure-Activity Relationship , beta-Alanine/pharmacokinetics
4.
Bioorg Med Chem Lett ; 29(15): 1974-1980, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31138472

ABSTRACT

A novel series of indazole/indole derivatives were discovered as glucagon receptor (GCGR) antagonists through scaffold hopping based on two literature leads: MK-0893 and LY-2409021. Further structure-activity relationship (SAR) exploration and optimization led to the discovery of multiple potent GCGR antagonists with excellent pharmacokinetic properties in mice and rats, including low systemic clearance, long elimination half-life, and good oral bioavailability. These potent GCGR antagonists could be used for potential treatment of type II diabetes.


Subject(s)
Indazoles/chemistry , Receptors, Glucagon/antagonists & inhibitors , Humans , Molecular Structure , Structure-Activity Relationship
5.
J Med Chem ; 61(22): 10276-10298, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30339387

ABSTRACT

A novel series of 6-benzhydryl-4-amino-quinolin-2-ones was discovered as cannabinoid type 1 receptor (CB1R) inverse agonists based on the high-throughput screening hit, compound 1a. Structure-activity relationships were studied to improve in vitro/in vivo pharmacology and restrict distribution to the peripheral circulation. We adopted several strategies such as increasing topological polar surface area, incorporating discrete polyethylene glycol side chains, and targeting P-glycoprotein (P-gp) to minimize access to the brain. Compound 6a is a P-gp substrate and a potent and highly selective CB1R inverse agonist, demonstrating excellent in vivo metabolic stability and a low brain to plasma ratio. However, brain receptor occupancy studies showed that compound 6a may accumulate in brain with repeat dosing. This was evidenced by compound 6a inhibiting food intake and inducing weight loss in diet-induced obese mice. Thus, a strategy based on P-gp efflux may not be adequate for peripheral restriction of the disclosed quinolinone series.


Subject(s)
Drug Inverse Agonism , Quinolones/chemistry , Quinolones/pharmacology , Receptor, Cannabinoid, CB1/agonists , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Conformation , Quinolones/metabolism , Quinolones/pharmacokinetics , Rats , Receptor, Cannabinoid, CB1/chemistry , Receptor, Cannabinoid, CB1/metabolism , Structure-Activity Relationship , Tissue Distribution
6.
Bioorg Med Chem Lett ; 28(21): 3446-3453, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30268701

ABSTRACT

A new series of (2S,3R,4R,5S,6R)-5-fluoro-6-(hydroxymethyl)-2-aryltetrahydro-2H-pyran-3,4-diols as dual inhibitors of sodium glucose co-transporter proteins (SGLTs) were disclosed. Two methods were developed to efficiently synthesize C5-fluoro-lactones 3 and 4, which are key intermediates to the C5-fluoro-hexose based C-aryl glucosides. Compound 2b demonstrated potent hSGLT1 and hSGLT2 inhibition (IC50 = 43 nM for SGLT1 and IC50 = 9 nM for SGLT2). It showed robust inhibition of blood glucose excursion in oral glucose tolerance test (OGTT) in Sprague Dawley (SD) rats and exerted pronounced antihyperglycemic effects in db/db mice and high-fat diet-fed ZDF rats when dosed orally at 10 mg/kg.


Subject(s)
Deoxyglucose/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/therapeutic use , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Administration, Oral , Animals , Blood Glucose/drug effects , Deoxyglucose/administration & dosage , Deoxyglucose/analogs & derivatives , Deoxyglucose/chemical synthesis , Drug Design , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Macaca fascicularis , Male , Mice , Microsomes, Liver/metabolism , Molecular Structure , Rats, Sprague-Dawley , Rats, Zucker , Sodium-Glucose Transporter 2/metabolism , Sodium-Glucose Transporter 2 Inhibitors/administration & dosage , Sodium-Glucose Transporter 2 Inhibitors/chemical synthesis , Sodium-Glucose Transporter 2 Inhibitors/chemistry , Structure-Activity Relationship
7.
J Pharmacol Exp Ther ; 365(3): 676-687, 2018 06.
Article in English | MEDLINE | ID: mdl-29674332

ABSTRACT

The sodium/glucose cotransporters (SGLT1 and SGLT2) transport glucose across the intestinal brush border and kidney tubule. Dual SGLT1/2 inhibition could reduce hyperglycemia more than SGLT2-selective inhibition in patients with type 2 diabetes. However, questions remain about altered gastrointestinal (GI) luminal glucose and tolerability, and this was evaluated in slc5a1-/- mice or with a potent dual inhibitor (compound 8; SGLT1 Ki = 1.5 ± 0.5 nM 100-fold greater potency than phlorizin; SGLT2 Ki = 0.4 ± 0.2 nM). 13C6-glucose uptake was quantified in slc5a1-/- mice and in isolated rat jejunum. Urinary glucose excretion (UGE), blood glucose (Sprague-Dawley rats), glucagon-like peptide 1 (GLP-1), and hemoglobin A1c (HbA1c) levels (Zucker diabetic fatty rats) were measured. Intestinal adaptation and rRNA gene sequencing was analyzed in C57Bl/6 mice. The blood 13C6-glucose area under the curve (AUC) was reduced in the absence of SGLT1 by 75% (245 ± 6 vs. 64 ± 6 mg/dl⋅h in wild-type vs. slc5a1-/- mice) and compound 8 inhibited its transport up to 50% in isolated rat jejunum. Compound 8 reduced glucose excursion more than SGLT2-selective inhibition (e.g., AUC = 129 ± 3 vs. 249 ± 5 mg/dl⋅h for 1 mg/kg compound 8 vs. dapagliflozin) with similar UGE but a lower renal glucose excretion threshold. In Zucker diabetic fatty rats, compound 8 decreased HbA1c and increased total GLP-1 without changes in jejunum SGLT1 expression, mucosal weight, or villus length. Overall, compound 8 (1 mg/kg for 6 days) did not increase cecal glucose concentrations or bacterial diversity in C57BL/6 mice. In conclusion, potent dual SGLT1/2 inhibition lowers blood glucose by reducing intestinal glucose absorption and the renal glucose threshold but minimally impacts the intestinal mucosa or luminal microbiota in chow-fed rodents.


Subject(s)
Blood Glucose/metabolism , Colon/drug effects , Colon/microbiology , Microbiota/drug effects , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2/metabolism , Animals , Biodiversity , Colon/metabolism , Male , Mice , Rats , Sodium-Glucose Transporter 2 Inhibitors/metabolism
8.
Bioorg Med Chem Lett ; 28(7): 1182-1187, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29523385

ABSTRACT

Synthesis and biological evaluation of benzocyclobutane-C-glycosides as potent and orally active SGLT1/SGLT2 dual inhibitors are described. Compound 19 showed high inhibitory potency at SGLT1 (IC50 = 45 nM), and excellent potency at SGLT2 (IC50 = 1 nM). It also displayed excellent PK profiles in mice, rats, dogs and monkeys (F = 78-107%). In SD rats, compound 19 treatments significantly reduced blood glucose levels in a dose-dependent manner. In ZDF rats, compound 19 displayed anti-hyperglycemic effect up to 24 h. Therefore, compound 19 may serve as valuable pharmacological tool, and potential use as a treatment for metabolic syndrome.


Subject(s)
Benzene Derivatives/pharmacology , Cyclobutanes/pharmacology , Glycosides/pharmacology , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors , Administration, Oral , Animals , Benzene Derivatives/administration & dosage , Benzene Derivatives/chemistry , Cyclobutanes/administration & dosage , Cyclobutanes/chemistry , Dogs , Dose-Response Relationship, Drug , Glycosides/administration & dosage , Glycosides/chemistry , Haplorhini , Humans , Mice , Molecular Structure , Rats , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 2/metabolism , Structure-Activity Relationship
9.
ACS Med Chem Lett ; 8(5): 560-565, 2017 May 11.
Article in English | MEDLINE | ID: mdl-28523111

ABSTRACT

We have discovered a novel series of tetrahydrobenzimidazoles 3 as TGR5 agonists. Initial structure-activity relationship studies with an assay that measured cAMP levels in murine enteroendocrine cells (STC-1 cells) led to the discovery of potent agonists with submicromolar EC50 values for mTGR5. Subsequent optimization through methylation of the 7-position of the core tetrahydrobenzimidazole ring resulted in the identification of potent agonists for both mTGR5 and hTGR5 (human enteroendocrine NCI-H716 cells). While the lead compounds displayed low to moderate exposure after oral dosing, they significantly reduced blood glucose levels in C57 BL/6 mice at 30 mg/kg and induced a 13-22% reduction in the area under the blood glucose curve (AUC)0-120 min in oral glucose tolerance tests (OGTT).

11.
Ann N Y Acad Sci ; 1358: 28-43, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26305874

ABSTRACT

The sodium glucose co-transporter 2 (SGLT2) inhibitor canagliflozin is a novel treatment option for adults with type 2 diabetes mellitus (T2DM). In patients with hyperglycemia, SGLT2 inhibition lowers plasma glucose levels by reducing the renal threshold for glucose (RTG ) and increasing urinary glucose excretion (UGE). Increased UGE is also associated with a mild osmotic diuresis and net caloric loss, which can lead to reductions in body weight and blood pressure (BP). After promising results from preclinical and phase I/II studies, the efficacy and safety of canagliflozin was evaluated in a comprehensive phase III development program in over 10,000 patients with T2DM on various background therapies. Canagliflozin improved glycemic control and provided reductions in body weight and BP versus placebo and active comparators in studies of up to 2 years' duration. Canagliflozin was generally well tolerated, with higher incidences of adverse events (AEs) related to the mechanism of action, including genital mycotic infections and AEs related to osmotic diuresis. Results from the preclinical and clinical studies led canagliflozin to be the first-in-class SGLT2 inhibitor approved in the United States, and support canagliflozin as a safe and effective therapeutic option across a broad range of patients with T2DM.


Subject(s)
Canagliflozin/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Canagliflozin/pharmacology , Humans , Hypoglycemic Agents/pharmacology
12.
PLoS One ; 9(9): e110069, 2014.
Article in English | MEDLINE | ID: mdl-25268802

ABSTRACT

INTRODUCTION: This randomized, double-blind, placebo-controlled, single and multiple ascending-dose study evaluated the pharmacodynamic effects and safety/tolerability of canagliflozin, a sodium glucose co-transporter 2 inhibitor, in patients with type 2 diabetes. METHODS: Patients (N = 116) discontinued their antihyperglycemic medications 2 weeks before randomization. Patients received canagliflozin 30, 100, 200, or 400 mg once daily or 300 mg twice daily, or placebo at 2 study centers in the United States and Germany, or canagliflozin 30 mg once daily or placebo at 1 study center in Korea, while maintaining an isocaloric diet for 2 weeks. On Days -1, 1, and 16, urinary glucose excretion (UGE), plasma glucose (PG), fasting PG (FPG), and insulin were measured. The renal threshold for glucose (RTG) was calculated from UGE, PG, and estimated glomerular filtration rate. Safety was evaluated based on adverse event (AE) reports, vital signs, electrocardiograms, clinical laboratory tests, and physical examinations. RESULTS: Canagliflozin increased UGE dose-dependently (,80-120 g/day with canagliflozin $100 mg), with increases maintained over the 14-day dosing period with each dose. Canagliflozin dose-dependently decreased RTG, with maximal reductions to ,4-5 mM (72-90 mg/dL). Canagliflozin also reduced FPG and 24-hour mean PG; glucose reductions were seen on Day 1 and maintained over 2 weeks. Plasma insulin reductions with canagliflozin were consistent with observed PG reductions. Canagliflozin also reduced body weight. AEs were transient, mild to moderate in intensity, and balanced across groups; 1 canagliflozin-treated female reported an episode of vaginal candidiasis. Canagliflozin did not cause hypoglycemia, consistent with the RTG values remaining above the hypoglycemia threshold. At Day 16, there were no clinically meaningful changes in urine volume, urine electrolyte excretion, renal function, or routine laboratory test values. CONCLUSIONS: Canagliflozin increased UGE and decreased RTG, leading to reductions in PG, insulin, and body weight, and was generally well tolerated in patients with type 2 diabetes. TRIAL REGISTRATION: ClinicalTrials.gov NCT00963768.


Subject(s)
Canagliflozin/pharmacokinetics , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacokinetics , Sodium-Glucose Transporter 2 Inhibitors , Adult , Blood Glucose/metabolism , Body Weight/drug effects , Canagliflozin/administration & dosage , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Fasting , Female , Glomerular Filtration Rate/drug effects , Glycosuria/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Insulin/blood , Male , Middle Aged , Sodium-Glucose Transporter 2/metabolism , Urinalysis
13.
PLoS One ; 9(8): e105638, 2014.
Article in English | MEDLINE | ID: mdl-25166023

ABSTRACT

INTRODUCTION: This randomized, double-blind, placebo-controlled, single and multiple ascending-dose study evaluated the pharmacodynamic effects and safety/tolerability of canagliflozin, a sodium glucose co-transporter 2 inhibitor, in patients with type 2 diabetes. METHODS: Patients (N = 116) discontinued their antihyperglycemic medications 2 weeks before randomization. Patients received canagliflozin 30, 100, 200, or 400 mg once daily or 300 mg twice daily, or placebo at 2 study centers in the United States and Germany, or canagliflozin 30 mg once daily or placebo at 1 study center in Korea, while maintaining an isocaloric diet for 2 weeks. On Days -1, 1, and 16, urinary glucose excretion (UGE), plasma glucose (PG), fasting PG (FPG), and insulin were measured. The renal threshold for glucose (RTG) was calculated from UGE, PG, and estimated glomerular filtration rate. Safety was evaluated based on adverse event (AE) reports, vital signs, electrocardiograms, clinical laboratory tests, and physical examinations. RESULTS: Canagliflozin increased UGE dose-dependently (∼80-120 g/day with canagliflozin ≥100 mg), with increases maintained over the 14-day dosing period with each dose. Canagliflozin dose-dependently decreased RTG, with maximal reductions to ∼4-5 mM (72-90 mg/dL). Canagliflozin also reduced FPG and 24-hour mean PG; glucose reductions were seen on Day 1 and maintained over 2 weeks. Plasma insulin reductions with canagliflozin were consistent with observed PG reductions. Canagliflozin also reduced body weight. AEs were transient, mild to moderate in intensity, and balanced across groups; 1 canagliflozin-treated female reported an episode of vaginal candidiasis. Canagliflozin did not cause hypoglycemia, consistent with the RTG values remaining above the hypoglycemia threshold. At Day 16, there were no clinically meaningful changes in urine volume, urine electrolyte excretion, renal function, or routine laboratory test values. CONCLUSIONS: Canagliflozin increased UGE and decreased RTG, leading to reductions in PG, insulin, and body weight, and was generally well tolerated in patients with type 2 diabetes. TRIAL REGISTRATION: ClinicalTrials.gov NCT00963768.


Subject(s)
Body Weight/drug effects , Diabetes Mellitus, Type 2/drug therapy , Glucosides/pharmacology , Hypoglycemic Agents/pharmacology , Kidney/drug effects , Thiophenes/pharmacology , Adult , Aged , Blood Glucose , Body Weight/physiology , Canagliflozin , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Double-Blind Method , Female , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Glucosides/adverse effects , Glucosides/therapeutic use , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use , Insulin/blood , Kidney/physiopathology , Male , Middle Aged , Thiophenes/adverse effects , Thiophenes/therapeutic use
14.
Bioorg Med Chem Lett ; 24(5): 1437-41, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24405703

ABSTRACT

Design, synthesis, and biological evaluation of pyridazine-based, 4-bicyclic heteroaryl-piperidine derivatives as potent stearoyl-CoA desaturase-1 (SCD1) inhibitors are described. In a chronic study of selected analog (3e) in Zucker fa/fa (ZF) rat, dose-dependent decrease of body weight gain and plasma fatty acid desaturation index (DI) in both C16 and C18 are also demonstrated. The results indicate that the plasma fatty acid DI may serve as an indicator for direct target engagement and biomarker for SCD1 inhibition.


Subject(s)
Bridged Bicyclo Compounds/chemistry , Enzyme Inhibitors/chemistry , Pyridazines/chemistry , Stearoyl-CoA Desaturase/antagonists & inhibitors , Administration, Oral , Animals , Body Weight/drug effects , Drug Design , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Half-Life , Humans , Mice , Microsomes, Liver/metabolism , Obesity/drug therapy , Pyridazines/pharmacokinetics , Pyridazines/pharmacology , Pyridazines/therapeutic use , Rats , Rats, Sprague-Dawley , Rats, Zucker , Stearoyl-CoA Desaturase/metabolism , Structure-Activity Relationship
16.
PLoS One ; 7(8): e42134, 2012.
Article in English | MEDLINE | ID: mdl-22870290

ABSTRACT

An increasing amount of evidence supports pleiotropic metabolic roles of the cannibinoid-1 receptor (CB1R) in peripheral tissues such as adipose, liver, skeletal muscle and pancreas. To further understand the metabolic consequences of specific blockade of CB1R function in peripheral tissues, we performed a 10-week-study with an anti-sense oligonucleotide directed against the CB1R in diet-induced obese (DIO) AKR/J mice. DIO AKR/J mice were treated with CB1R ASO Isis-414930 (6.25, 12.5 and 25 mg/kg/week) or control ASO Isis-141923 (25 mg/kg/week) via intraperitoneal injection for 10 weeks. At the end of the treatment, CB1R mRNA from the 25 mg/kg/week CB1R ASO group in the epididymal fat and kidney was decreased by 81% and 63%, respectively. Body weight gain was decreased in a dose-dependent fashion, significantly different in the 25 mg/kg/week CB1R ASO group (46.1±1.0 g vs veh, 51.2±0.9 g, p<0.05). Body fat mass was reduced in parallel with attenuated body weight gain. CB1R ASO treatment led to decreased fed glucose level (at week 8, 25 mg/kg/week group, 145±4 mg/dL vs veh, 195±10 mg/dL, p<0.05). Moreover, CB1R ASO treatment dose-dependently improved glucose excursion during an oral glucose tolerance test, whereas control ASO exerted no effect. Liver steatosis was also decreased upon CB1R ASO treatment. At the end of the study, plasma insulin and leptin levels were significantly reduced by 25 mg/kg/week CB1R ASO treatment. SREBP1 mRNA expression was decreased in both epididymal fat and liver. G6PC and fatty acid translocase/CD36 mRNA levels were also reduced in the liver. In summary, CB1R ASO treatment in DIO AKR/J mice led to improved insulin sensitivity and glucose homeostasis. The beneficial effects of CB1R ASO treatment strongly support the notion that selective inhibition of the peripheral CB1R, without blockade of central CB1R, may serve as an effective approach for treating type II diabetes, obesity and the metabolic syndrome.


Subject(s)
Diet/adverse effects , Obesity/drug therapy , Oligodeoxyribonucleotides, Antisense/pharmacology , RNA, Messenger/antagonists & inhibitors , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , Dose-Response Relationship, Drug , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Female , Glucose/genetics , Glucose/metabolism , Insulin/genetics , Insulin/metabolism , Liver/metabolism , Liver/pathology , Male , Metabolic Syndrome/chemically induced , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Mice , Mice, Inbred AKR , Obesity/chemically induced , Obesity/genetics , Obesity/metabolism , Organ Specificity/drug effects , Organ Specificity/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
17.
Int J Clin Pharmacol Ther ; 50(8): 584-94, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22578199

ABSTRACT

OBJECTIVE: To assess and validate the application of a non-radioactive assay for cholesteryl ester transfer protein (CETP) activity in clinical samples. DESIGN AND METHODS: In this Phase 0 study, CETP activity was measured following addition of the CETP inhibitor JNJ-28545595 to plasma samples from normolipidemic and three subgroups of dyslipidemic subjects with differing lipid profiles. RESULTS: CETP activity was elevated in plasma samples from dyslipidemic subjects compared to normolipidemic subjects. Increased triglyceride levels correlated with decreased CETP inhibition. The assay was found to have good analytical precision and high throughput potential as required for clinical trial sample analysis. CONCLUSIONS: The results demonstrate that pharmacological inhibition of CETP is affected by the dyslipidemic nature of plasma samples. In addition, since the optimal degree of CETP inhibition for maximal cardiovascular benefit in patients is not known, this assay may be used to help define optimal dosing of CETP inhibitors.


Subject(s)
Biological Assay/methods , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Dyslipidemias/blood , Lipids/blood , Adult , Aged , Cholesterol Ester Transfer Proteins/blood , Dose-Response Relationship, Drug , Female , Humans , In Vitro Techniques , Inhibitory Concentration 50 , Male , Middle Aged , Triglycerides/blood
18.
J Lipid Res ; 53(6): 1106-16, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22493088

ABSTRACT

Diacylglycerol acyltransferase (DGAT) catalyzes the final step in triglyceride (TG) synthesis. There are two isoforms, DGAT1 and DGAT2, with distinct protein sequences and potentially different physiological functions. To date, the ability to determine clear functional differences between DGAT1 and DGAT2, especially with respect to hepatic TG synthesis, has been elusive. To dissect the roles of these two key enzymes, we pretreated HepG2 hepatoma cells with (13)C(3)-D(5)-glycerol or (13)C(18)-oleic acid, and profiled the major isotope-labeled TG species by liquid chromatography tandem mass spectrometry. Selective DGAT1 and DGAT2 inhibitors demonstrated that (13)C(3)-D(5)-glycerol-incorporated TG synthesis was mediated by DGAT2, not DGAT1. Conversely, (13)C(18)-oleoyl-incorporated TG synthesis was predominantly mediated by DGAT1. To trace hepatic TG synthesis and VLDL triglyceride (VLDL-TG) secretion in vivo, we administered D(5)-glycerol to mice and measured plasma levels of D(5)-glycerol-incorporated TG. Treatment with an antisense oligonucleotide (ASO) to DGAT2 led to a significant reduction in D(5)-glycerol incorporation into VLDL-TG. In contrast, the DGAT2 ASO had no effect on the incorporation of exogenously administered (13)C(18)-oleic acid into VLDL-TG. Thus, our results indicate that DGAT1 and DGAT2 mediate distinct hepatic functions: DGAT2 is primarily responsible for incorporating endogenously synthesized FAs into TG, whereas DGAT1 plays a greater role in esterifying exogenous FAs to glycerol.


Subject(s)
Diacylglycerol O-Acyltransferase/metabolism , Enzyme Assays/methods , Glycerol/metabolism , Liver/enzymology , Oleic Acid/metabolism , Animals , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/genetics , Enzyme Inhibitors/pharmacology , Esterification/drug effects , Fatty Acids/biosynthesis , Fatty Acids/metabolism , Hep G2 Cells , Humans , Isotope Labeling , Lipoproteins, VLDL/metabolism , Male , Mice , Oligonucleotides, Antisense/genetics , Triglycerides/biosynthesis
19.
PLoS One ; 7(2): e30555, 2012.
Article in English | MEDLINE | ID: mdl-22355316

ABSTRACT

BACKGROUND: Canagliflozin is a sodium glucose co-transporter (SGLT) 2 inhibitor in clinical development for the treatment of type 2 diabetes mellitus (T2DM). METHODS: (14)C-alpha-methylglucoside uptake in Chinese hamster ovary-K cells expressing human, rat, or mouse SGLT2 or SGLT1; (3)H-2-deoxy-d-glucose uptake in L6 myoblasts; and 2-electrode voltage clamp recording of oocytes expressing human SGLT3 were analyzed. Graded glucose infusions were performed to determine rate of urinary glucose excretion (UGE) at different blood glucose (BG) concentrations and the renal threshold for glucose excretion (RT(G)) in vehicle or canagliflozin-treated Zucker diabetic fatty (ZDF) rats. This study aimed to characterize the pharmacodynamic effects of canagliflozin in vitro and in preclinical models of T2DM and obesity. RESULTS: Treatment with canagliflozin 1 mg/kg lowered RT(G) from 415±12 mg/dl to 94±10 mg/dl in ZDF rats while maintaining a threshold relationship between BG and UGE with virtually no UGE observed when BG was below RT(G). Canagliflozin dose-dependently decreased BG concentrations in db/db mice treated acutely. In ZDF rats treated for 4 weeks, canagliflozin decreased glycated hemoglobin (HbA1c) and improved measures of insulin secretion. In obese animal models, canagliflozin increased UGE and decreased BG, body weight gain, epididymal fat, liver weight, and the respiratory exchange ratio. CONCLUSIONS: Canagliflozin lowered RT(G) and increased UGE, improved glycemic control and beta-cell function in rodent models of T2DM, and reduced body weight gain in rodent models of obesity.


Subject(s)
Blood Glucose/metabolism , Body Weight/drug effects , Diabetes Mellitus, Experimental/drug therapy , Glucosides/therapeutic use , Hyperglycemia/drug therapy , Kidney/physiopathology , Thiophenes/therapeutic use , Animals , CHO Cells , Canagliflozin , Cells, Cultured , Cricetinae , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Glucose Tolerance Test , Humans , Hyperglycemia/metabolism , Hyperglycemia/pathology , Kidney/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Rats , Rats, Zucker , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transport Proteins/metabolism , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 2/genetics , Sodium-Glucose Transporter 2/metabolism , Sodium-Glucose Transporter 2 Inhibitors , Weight Gain/drug effects
20.
Bioorg Med Chem Lett ; 22(4): 1750-5, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22264481

ABSTRACT

The design and synthesis of a second generation GPR119-agonist clinical candidate for the treatment of diabetes is described. Compound 16 (APD597, JNJ-38431055) was selected for preclinical development based on a good balance between agonist potency, intrinsic activity and in particular on its good solubility and reduced drug-drug interaction potential. In addition, extensive in vivo studies showed a more favorable metabolic profile that may avoid the generation of long lasting metabolites with the potential to accumulate in clinical studies.


Subject(s)
Drug Discovery , Hypoglycemic Agents/chemistry , Piperidines/chemistry , Piperidines/pharmacokinetics , Pyridines/chemistry , Pyridines/pharmacokinetics , Receptors, G-Protein-Coupled/agonists , Animals , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Humans , Hypoglycemic Agents/pharmacokinetics , Mice , Mice, Inbred C57BL , Molecular Structure , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...