WO2010080333A1 - Arylcyclopropylacetamide derivatives useful as glucokinase activators - Google Patents

Arylcyclopropylacetamide derivatives useful as glucokinase activators Download PDF

Info

Publication number
WO2010080333A1
WO2010080333A1 PCT/US2009/067603 US2009067603W WO2010080333A1 WO 2010080333 A1 WO2010080333 A1 WO 2010080333A1 US 2009067603 W US2009067603 W US 2009067603W WO 2010080333 A1 WO2010080333 A1 WO 2010080333A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutically acceptable
compound
diabetes
acceptable salt
solution
Prior art date
Application number
PCT/US2009/067603
Other languages
French (fr)
Inventor
Ana Belen Bueno Melendo
Francisco Javier Agejas-Chicharro
Original Assignee
Eli Lilly And Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to UAA201107562A priority Critical patent/UA104742C2/en
Priority to CN200980150686XA priority patent/CN102256952B/en
Priority to BRPI0923183-8A priority patent/BRPI0923183B1/en
Priority to JP2011542276A priority patent/JP5497064B2/en
Priority to PL09768316T priority patent/PL2379517T3/en
Priority to EA201170845A priority patent/EA019055B1/en
Priority to SI200930713T priority patent/SI2379517T1/en
Priority to ES09768316T priority patent/ES2435242T3/en
Priority to MX2011006434A priority patent/MX2011006434A/en
Priority to AU2009335931A priority patent/AU2009335931B2/en
Application filed by Eli Lilly And Company filed Critical Eli Lilly And Company
Priority to EP09768316.3A priority patent/EP2379517B1/en
Priority to NZ592930A priority patent/NZ592930A/en
Priority to SG2011044823A priority patent/SG172256A1/en
Priority to CA2746746A priority patent/CA2746746C/en
Priority to KR1020117014073A priority patent/KR101290437B1/en
Priority to DK09768316.3T priority patent/DK2379517T3/en
Publication of WO2010080333A1 publication Critical patent/WO2010080333A1/en
Priority to ZA2011/03944A priority patent/ZA201103944B/en
Priority to IL213210A priority patent/IL213210A/en
Priority to TN2011000294A priority patent/TN2011000294A1/en
Priority to MA33945A priority patent/MA32902B1/en
Priority to HRP20130983AT priority patent/HRP20130983T1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/38Nitrogen atoms
    • C07D277/44Acylated amino or imino radicals
    • C07D277/46Acylated amino or imino radicals by carboxylic acids, or sulfur or nitrogen analogues thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • Diabetes is a progressive disease which adversely affects both longevity and quality of life.
  • Existing oral therapies either alone or in combination, do not exhibit 5 adequate or sustained glucose lowering efficacy in patients with diabetes. Consequently, there is an unmet need for improved therapies for patients with diabetes.
  • GKAs Glucokinase activators
  • GK Glucokinase
  • the compounds of the present invention have been found to activate glucokinase 20 both in vitro and in vivo.
  • the compounds of the present invention have been found to exhibit improved potency over existing GKAs.
  • the compounds of the present invention have been found to exhibit limited blood brain barrier permeability.
  • the present invention is directed to compounds which activate glucokinase, pharmaceutical compositions containing them as an active ingredient, methods for the 25 treatment of disorders associated with glucokinase dysfunction, and to their use for the treatment of diabetes, in particular Type II diabetes.
  • the present invention provides a compound of the formula: or a pharmaceutically acceptable salt thereof.
  • a compound of the present invention has two stereocenters (*) and thus four possible stereoisomers. It is intended that each stereoisomer and racemic or diastereomeric mixtures, whether pure or partially pure, are included within the scope of the invention.
  • a preferred stereoisomer of a compound of the present invention has the structural formula:
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable diluent or carrier.
  • the present invention provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in therapy.
  • the present invention also provides a compound of Formula I, or a pharmaceutically acceptable salt thereof for use in the treatment of diabetes, in particular type II diabetes.
  • a compound of Formula I, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of diabetes, in particular Type II diabetes.
  • the present invention provides a method for the treatment of diabetes, which comprises administering an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, to a human being or animal in need thereof.
  • the present invention also provides a method for the treatment of Type II diabetes, which comprises administering an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, to a human being or animal in need thereof.
  • the present invention provides a pharmaceutical composition for use in therapy comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof.
  • the present invention provides a pharmaceutical composition for use in diabetes, in particular type II diabetes, comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof.
  • the term "pharmaceutically acceptable salt” refers to salts of a compound of the present invention which are substantially non-toxic to living organisms. Such salts and common methodology for preparing them are well known in the art. See, e.g., P. Stahl, et ah, Handbook of Pharmaceutical Salts: Properties Selection and Use, (VCHA/Wiley-VCH, 2002); and J. Pharm. Sci. 66, 2-19 (1977).
  • a preferred pharmaceutically acceptable salt is hydrochloride.
  • the compounds of the present invention are preferably formulated as pharmaceutical compositions administered by a variety of routes. Most preferably, such compositions are for oral administration. Such pharmaceutical compositions and processes for preparing same are well known in the art. See, e.g., Remington: The Science and Practice of Pharmacy (A, Gennaro, et ah, eds., 19 th ed., Mack Publishing Co., 1995).
  • the present compounds are administered in combination with one or more active substances.
  • active substances include for example metformin.
  • Administration in combination includes simultaneous, sequential or separate administration.
  • the compound names for the following example are generated using AutoNom 2000.
  • aqueous phases are back-extracted with ethyl acetate (2 x 500 ml), and the organic layers are combined, dried over magnesium sulfate, and filtered.
  • This crude hydrazone solution ( ⁇ 2.1 L, assumed to contain 363 g of hydrozone intermediate) is stirred well while triethylamine (100 mL, 890 mmol) is added slowly. The resulting solution is left to stand overnight, during which time some solid precipitates.
  • the mixture is diluted with ethyl acetate to a volume of 3 L, affording a solution, which is washed with 1 L of water, followed by two 500 mL portions of water combined with saturated aqueous sodium chloride solution as necessary to break up any emulsions.
  • the resulting slurry is then stirred at 50 0 C for 1 hour, during which time a solution forms.
  • Methanol is removed under vacuum, and 1 L of ethylacetate is added.
  • the resulting mixture is acidified by the addition of approximately 550 mL of 5% aqueous hydrochloric acid, and the two layers are separated.
  • the aqueous layer is then extracted with two 500 mL portions of ethyl acetate.
  • the organic phases are combined, washed with 500 mL of saturated aqueous sodium chloride solution, dried over magnesium sulfate, filtered, and concentrated to afford a moist pale yellow solid. This material is then dissolved in 1 L of methanol. Water (1 L) is then added to the stirred solution over the course of 1.5 hours.
  • the enantiomeric excess of the acid is determined to be 97.7% by comparison of the integrals for the two peaks corresponding to the enantiomers as separated by chiral chromatography on an AD-H column (150 mm) eluted at 35°C with 10% ethanol in hexanes containing 0.05% trifluoroacetic acid.
  • Oxalyl chloride (146.89 mL, 1.69 mol) is added over 15 min to a stirred solution of (lR,2S)-2-cyclohexyl-l-(4-cyclopropanesulfonyl-phenyl)-cyclopropanecarboxylic acid (295.00 g, 0.847 mol) in 10 L of anhydrous dichloromethane under an inert atmosphere.
  • Dimethylformamide (654.61 ⁇ L, 8.5 mmol) is then added at once, and the resulting solution is stirred overnight. Volatiles are then removed under vacuum at 40 0 C to afford an oil, which is dissolved in 3 L of anhydrous dichloromethane.
  • the resulting mixture is stirred for a few minutes, and then the two layers are separated.
  • the aqueous layer is extracted with 1 L dichloromethane, and the dichloromethane solutions are combined, dried over MgSO 4 , filtered, and concentrated under vacuum at 40 0 C.
  • the resulting oil (556 g) is applied to silica gel plugs as a dichloromethane solution. Elution of the plugs with 1: 12:7 2M ammonia in methanol/ methyl t-butyl ether /heptane, followed by 1: 19 2M ammonia (in methanol)/ethyl acetate affords a brown foam (351 g).
  • the human islet GK isoform is expressed in E.coli as (His)6 -tagged fusion protein and purified with metal chelate affinity chromatography, see e.g. Tiedge et al., Biochem. Biophys. Acta 1337, 175-190, 1997. After purification the enzyme is stored in aliquots at concentration 0.8 mg/ml in 25 mM sodium phosphate, 150 mM sodium chloride, 100 mM imidazole, 1 mM dithiothreitol, 50 % glycerol at -80 0 C. The assay is performed in flat bottom 96-well plates in a final incubation volume of lOO ⁇ L.
  • the incubation mixture consists of 25 mM 4-(2-hydroxyethyl)-l-piperazineethanesulfonic acid (HEPES) (pH7.4), 50 mM potassiumchloride, 2.5 mM magnesiumchloride, 2 mM dithiothreitol, 4 U/ml glucose-6-phosphate dehydrogenase from Leuconostoc mesenteroides, 5 mM ATP, 1 mM NAD and a set concentration of glucose. Test compounds are dissolved in dimethylsulfoxide and then added to the reaction mixture giving the final dimethylsulfoxide concentration of 10%. The reaction is initiated by addition of 20 ⁇ L GK and run for 20 min at 37°C. The amount of formed NADH is measured as an increase in absorbance at 340 nm using a microplate reader. Absorbance values are used for EC50 calculations.
  • HEPES 4-(2-hydroxyethyl)-l-piperazineethanesulfonic acid
  • Rat insulinoma INS-IE cells are maintained at 37 0 C, 5% CO 2 , 95% humidity in 1640 medium supplemented with 11 mM glucose, 5% Fetal Bovine Serum, 50 ⁇ M 2- Mercaptoethanol, 1 mM pyruvate, 10 mM HEPES and antibiotics. Prior to assay, cells are trypsinized, pelleted by centrifugation and seeded into 96-well tissue culture assay plates at the density of 30,000 cells/well. Cells are allowed to attach and incubated for 48 hours at 37 0 C, 5% CO 2 .
  • EBSS Earle's Balanced Salt Solution
  • BSA Bovine Serum Albumin
  • EDTA ethylenediaminetetraacetic acid
  • Example 1 decreases plasma glucose in a dose-dependent manner at both fasted and postprandial glucose levels.
  • a maximal lowering of glucose AUC versus the untreated control group is observed with the high dose (30 mg/kg) and represents a 42% decrease.
  • Interpolation of the data showed that a 20% glucose AUC decrease occurs at an average compound concentration of 99 ng/ml (179 nM) in plasma, corresponding to a 6.9 mg/kg compound dose.
  • the compounds of the present invention are shown to activate GK in vivo.
  • a stock compound solution is prepared in dimethylsulfoxide at 10 mM.
  • a dose solution is then prepared at ImM by diluting 100 ⁇ L of the stock with 900 ⁇ L of propylene glycol.
  • the dose is administered as an IV bolus (2.2 mL/kg) via the tail vein into six male CF-I mice (approximately 23 g) for a target dose of 2.17 ⁇ mole/kg.
  • Mice are euthanized using both CO 2 and cervical dislocation. Three mice are sacrificed 5 minutes post dose and three 60 minutes post dose. Blood is collected from each animal via cardiac puncture, and plasma is prepared using sodium EDTA, transferred into polypropylene sample tubes and immediately frozen using dry ice.
  • Plasma samples are prepared for analysis by precipitation of protein using two parts of extraction sovent (10% tetrahydrofuran in acetonitrile) to one part plasma and mixing with a vortex mixer. For brain tissue, it is assumed that 1 mg brain tissue ⁇ 1 ⁇ L volume and two parts of extraction solvent are added to one part tissue. The samples are immediately homogenized using an ultrasoinc cell dismemberator. Calibration standards are prepared by spiking known concentrations of compound into blank mouse plasma and then treated as plasma samples. All samples are centrifuged at 6000 RCF for 5 minutes. An aliquot of supernatant from each sample is transferred to a polypropylene 96 well plate and sealed for analysis by LC-MS/MS.
  • MS/MS is effected using a Sciex API 4000 triple quadrupole mass spectrometer equipped with a turbo ion spray source.
  • High Performance Liquid Chromatogaphy is effected using a Phenomenex Hyrdro RP analytical column (10Ox 2.0 mm, 4 ⁇ ) heated to 50 0 C and operated at a constant flow rate of 0.6 mL/minute.
  • a mobile phase gradient is utilized consisting of an initial mobile phase of 60:40 5 mM aqueous ammonium formate:5mM ammonium formate in methanol with a hold time of 1 minute followed by a linear 2 minute gradient to 10:90 5 mM aqueous ammonium formate: 5 mM ammonium formate in methanol with a final hold time of 1 minute.
  • Column effluent is diverted to waste from 0-2.8 minutes and then directed into the mass spectrometer from 2.8-4.0 minutes MS/MS transitions monitored are 560/84.
  • Quantitation of compound in test samples is achieved by comparing peak area values to a quadratic equation weighted 1/x 2 derived from the nominal concentrations of the calibration standards and their respective peak areas.
  • Upper and Lower Limits of Quantitation are determined by the back calculated recoveries of calibration standards that exceeded +/- 20% of theory. Brain tissue concentrations are corrected for plasma contribution using a literature factor of 16 ⁇ L of plasma/gm mouse brain.
  • Example 1 The in vivo blood brain barrier permeability of Example 1 resulted in a mean brain/plasma ratio of 0.17 five minutes post dose and a mean total brain level of 0.539 nmol/g at that time.
  • the compounds of the present invention have been shown to have limited blood brain barrier permeability and so provides limited potential for severe hypoglycaemia.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Diabetes (AREA)
  • General Chemical & Material Sciences (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Biomedical Technology (AREA)
  • Endocrinology (AREA)
  • Psychiatry (AREA)
  • Emergency Medicine (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

A compound of formula (A); and pharmaceutical compositions for the treatment of diabetes.

Description

ARYLCYCLOPROPYLACETAMIDE DERIVATIVES USEFUL AS GLUCOKINASE ACTIVATORS
Diabetes is a progressive disease which adversely affects both longevity and quality of life. Existing oral therapies, either alone or in combination, do not exhibit 5 adequate or sustained glucose lowering efficacy in patients with diabetes. Consequently, there is an unmet need for improved therapies for patients with diabetes.
Glucokinase activators (GKAs) represent a class of glucose-lowering agents which primarily act to lower blood glucose through modulatory actions in the pancreatic β-cells and the liver. A number of synthetic GKAs have been disclosed for the treatment 10 of diabetes, for example those disclosed in WO 04/063179. There remains a need for alternative GKAs as therapy for patients with diabetes.
It has been shown that Glucokinase (GK) is critical for mediation of glucose sensing in neurons. GK activation in the hypothalamus dampens the counterregulatory response to insulin-induced hypoglycemia. Thus, activation of GK in the brain with GKA 15 may produce an increased risk for hypoglycemia by decreasing secretion of epinephrine, norepinephrine, and glucagon levels at low glucose levels. GKA compounds with limited blood brain barrier permeability would have a lower potential for producing severe hypoglycemia.
The compounds of the present invention have been found to activate glucokinase 20 both in vitro and in vivo. The compounds of the present invention have been found to exhibit improved potency over existing GKAs. The compounds of the present invention have been found to exhibit limited blood brain barrier permeability.
The present invention is directed to compounds which activate glucokinase, pharmaceutical compositions containing them as an active ingredient, methods for the 25 treatment of disorders associated with glucokinase dysfunction, and to their use for the treatment of diabetes, in particular Type II diabetes.
The present invention provides a compound of the formula:
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof.
A compound of the present invention has two stereocenters (*) and thus four possible stereoisomers. It is intended that each stereoisomer and racemic or diastereomeric mixtures, whether pure or partially pure, are included within the scope of the invention.
A preferred stereoisomer of a compound of the present invention has the structural formula:
Figure imgf000003_0002
The present invention provides a pharmaceutical composition comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable diluent or carrier.
The present invention provides a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in therapy. The present invention also provides a compound of Formula I, or a pharmaceutically acceptable salt thereof for use in the treatment of diabetes, in particular type II diabetes. In another aspect of the present invention, there is provided the use of a compound of Formula I, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of diabetes, in particular Type II diabetes. The present invention provides a method for the treatment of diabetes, which comprises administering an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, to a human being or animal in need thereof. The present invention also provides a method for the treatment of Type II diabetes, which comprises administering an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, to a human being or animal in need thereof.
The present invention provides a pharmaceutical composition for use in therapy comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof. The present invention provides a pharmaceutical composition for use in diabetes, in particular type II diabetes, comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof.
As used herein the term "pharmaceutically acceptable salt" refers to salts of a compound of the present invention which are substantially non-toxic to living organisms. Such salts and common methodology for preparing them are well known in the art. See, e.g., P. Stahl, et ah, Handbook of Pharmaceutical Salts: Properties Selection and Use, (VCHA/Wiley-VCH, 2002); and J. Pharm. Sci. 66, 2-19 (1977). A preferred pharmaceutically acceptable salt is hydrochloride. The compounds of the present invention are preferably formulated as pharmaceutical compositions administered by a variety of routes. Most preferably, such compositions are for oral administration. Such pharmaceutical compositions and processes for preparing same are well known in the art. See, e.g., Remington: The Science and Practice of Pharmacy (A, Gennaro, et ah, eds., 19th ed., Mack Publishing Co., 1995).
In a further aspect of the invention the present compounds are administered in combination with one or more active substances. Such active substances include for example metformin.
Administration in combination includes simultaneous, sequential or separate administration.
The compound names for the following example are generated using AutoNom 2000.
General Procedures: All water-or air-sensitive reactions are conducted in dry solvents under an inert atmosphere. Mass spectra (MS) are obtained on an Agilent 1100 MSD spectrometer operating in electrospray mode. Optical rotations are obtained in chloroform on a JASCO DIP-370 digital polarimeter at 200C with a sodium D line.
Example 1 : (lR,2S)-2-Cyclohexyl-l-(4-cyclopropanesulfonyl-phenyl)- cyclopropanecarboxylic acid [5-(2-pyrrolidin-l-yl-ethylsulfanyl)-thiazol-2-yl]-amide
Figure imgf000005_0001
A) (4-Cyclopropanesulfonyl-phenyl)-diazo-acetic acid ethyl ester
Figure imgf000005_0002
A mixture of (4-cyclopropanesulfonyl-phenyl)-oxo-acetic acid ethyl ester (250 g,
806 mmol) and p-toluenesulfonyl hydrazide (187g, 984 mmol) in 1.5 L of ethanol is stirred at room temperature until a light yellow solution is obtained. Concentrated hydrochloric acid (20 mL, 233 mmol) is then added, and the resulting mixture is heated at reflux for 3.5 h. Volatiles are removed to provide a clear light yellow oil, which is dissolved in 1.5 L of ethyl acetate. This solution is then washed with 1 L of saturated aqueous sodium bicarbonate solution, followed by 1 L of saturated aqueous sodium chloride solution. The aqueous phases are back-extracted with ethyl acetate (2 x 500 ml), and the organic layers are combined, dried over magnesium sulfate, and filtered. This crude hydrazone solution (~ 2.1 L, assumed to contain 363 g of hydrozone intermediate) is stirred well while triethylamine (100 mL, 890 mmol) is added slowly. The resulting solution is left to stand overnight, during which time some solid precipitates. The mixture is diluted with ethyl acetate to a volume of 3 L, affording a solution, which is washed with 1 L of water, followed by two 500 mL portions of water combined with saturated aqueous sodium chloride solution as necessary to break up any emulsions. The resulting organic phase is then dried over magnesium sulfate, filtered, and concentrated to afford a damp solid, which is triturated with methyl t-butyl ether. The resulting slurry is filtered to afford a light yellow solid, which is dried under vacuum to afford 155 g of the title compound. The filtrate is concentrated to an oil, which is triturated as above until a free- flowing solid is obtained. This solid is isolated by filtration and dried to afford an additional 10 g of the title compound. LCMS (m/e): 295 (M+H).
B) (lR,2S)-2-Cyclohexyl-l-(4-cyclopropanesulfonyl-phenyl)-cyclopropanecarboxylic acid
Figure imgf000006_0001
To a solution of vinylcyclohexane (300 mL, 2.72 mol) in 150 mL of anhydrous dichloromethane maintained at 25-300C under an inert atmosphere is added a solution of tetrakis[N-phthaloyl-(R)-tert-leucinato]dirhodium bis(ethylacetate) adduct (120 mg, 84 μmol) in vinylcyclohexane (40 mL) dropwise, while portions of (4-cyclopropanesulfonyl- phenyl)-diazo-acetic acid ethyl ester (169.40 g, 575.5 mmol) are added. The addition rates are adjusted to maintain an internal temperature of 400C. The addition is complete after approximately 1.5 hours, and the reaction mixture is stirred for an additional 2 hours at 300C. Volatiles are then removed under vacuum to afford crude (lR,2S)-2-cyclohexyl-l- (4-cyclopropanesulfonyl-phenyl)-cyclopropanecarboxylic acid ethyl ester as a viscous brown oil (218 g, 579 mmol) which is dissolved in 1.1 L of methanol to afford a yellow- brown solution, to which a 5 N aqueous sodium hydroxide solution (500 mL, 2.5 mmol) is added slowly. The resulting slurry is then stirred at 500C for 1 hour, during which time a solution forms. Methanol is removed under vacuum, and 1 L of ethylacetate is added. The resulting mixture is acidified by the addition of approximately 550 mL of 5% aqueous hydrochloric acid, and the two layers are separated. The aqueous layer is then extracted with two 500 mL portions of ethyl acetate. The organic phases are combined, washed with 500 mL of saturated aqueous sodium chloride solution, dried over magnesium sulfate, filtered, and concentrated to afford a moist pale yellow solid. This material is then dissolved in 1 L of methanol. Water (1 L) is then added to the stirred solution over the course of 1.5 hours. The resulting slurry is stirred at room temperature for 30 minutes, and then filtered. The filter pad is washed with 1 : 1 methanol/water, and dried to afford the title compound as pale yellow crystals (166 g). MS exact mass calculated 349.14735; found 349.14679 (Agilent 1100 LC-TOF using electrospray ionization); [a]^2 = - 31°.
The enantiomeric excess of the acid is determined to be 97.7% by comparison of the integrals for the two peaks corresponding to the enantiomers as separated by chiral chromatography on an AD-H column (150 mm) eluted at 35°C with 10% ethanol in hexanes containing 0.05% trifluoroacetic acid.
C) 5-(2-Pyrrolidin-l-yl-ethylsulfanyl)-thiazol-2-ylamine
Figure imgf000007_0001
Thiirane (550 mL, 9.2 mol ) is added to a mixture of pyrrolidine (543 mL, 6.57 mol) in 2.5 L of anhydrous dioxane under an inert atmosphere. The temperature rises slowly, and the reaction mixture is cooled in an ice bath when the internal temperature reaches 54°C. Once the temperature has dropped to 45°C, the cooling bath is removed and the reaction mixture is heated to 600C. After 3 hours, the mixture is cooled to room temperature and concentrated under vacuum. The residue is then distilled at 6 mm Hg, and a fraction boiling at 500C is collected to afford 2-pyrrolidin-l-yl-ethanethiol as a colorless oil (643 g). MS (m/e): 132 (M+H).
Sodium bicarbonate ((1.232 kg, 14.7 mol) is added slowly in portions to a mixture of 5-bromo-thiazol-2-ylamine hydrobromide (1.53 Kg, 5.87 mol) in 7.5 L of isopropanol. 2-Pyrrolidin-l-yl-ethanethiol (1.060 Kg, 8.07 mol) is then added over 15 min, and the resulting mixture is stirred at 600C for 96 h. The temperature is increased to 700C for 1 h, and then the mixture is cooled to room temperature. Most of the isopropanol is removed under vacuum, and the residue is taken up in 4 L of an isopropanol/chloroform solution (1 :9). Saturated aqueous sodium bicarbonate (4 L) is added, and the resulting mixture is stirred for 30 minutes. The layers are separated and the aqueous phase is extracted with three 4 L portions of an isopropanol/chloroform solution (1 :9). The organic layers are combined, dried over sodium sulfate, filtered, and concentrated under vacuum. The resulting residue is triturated with 3 L of diethylether, and filtered off to give a first portion of the title compound as a pale-yellow solid (410 g). The filtrate is concentrated to an orange solid, which is triturated with 2 L of diethylether, and isolated as a beige solid by filtration. This solid is then dissolved in 2 L of methanol, and the solution is heated at 45 0C for 30 min. Upon cooling to room temperature, a solid is formed. This material is isolated by filtration, triturated with diethyl ether as above, and dried under vacuum to afford an additional 310 g of the title compound. MS (m/e): 230 [M+H]
D) (lR,2S)-2-Cyclohexyl-l-(4-cyclopropanesulfonyl-phenyl)-cyclopropanecarboxylic acid [5-(2-pyrrolidin-l-yl-ethylsulfanyl)-thiazol-2-yl]-amide
Oxalyl chloride (146.89 mL, 1.69 mol) is added over 15 min to a stirred solution of (lR,2S)-2-cyclohexyl-l-(4-cyclopropanesulfonyl-phenyl)-cyclopropanecarboxylic acid (295.00 g, 0.847 mol) in 10 L of anhydrous dichloromethane under an inert atmosphere. Dimethylformamide (654.61 μL, 8.5 mmol) is then added at once, and the resulting solution is stirred overnight. Volatiles are then removed under vacuum at 400C to afford an oil, which is dissolved in 3 L of anhydrous dichloromethane. An inert atmosphere is reestablished, and the solution is cooled to <5°C. Triethylamine (177 mL 1.27 mol) is then added dropwise over 20 minutes, leaving a dark solution. Sodium Sulfate (120.25 g, 0.847 mol) is added, followed by 5-(2-pyrrolidin-l-yl-ethylsulfanyl)-thiazol-2-ylamine (213.60 g 0.931 mole). The internal temperature rises to 200C. The reaction mixture is stirred for 10 min in the cold, and is then allowed to warm to room temperature. After being stirred overnight, the reaction mixture is poured onto 3 L of water. The resulting mixture is stirred for a few minutes, and then the two layers are separated. The aqueous layer is extracted with 1 L dichloromethane, and the dichloromethane solutions are combined, dried over MgSO4, filtered, and concentrated under vacuum at 400C. The resulting oil (556 g) is applied to silica gel plugs as a dichloromethane solution. Elution of the plugs with 1: 12:7 2M ammonia in methanol/ methyl t-butyl ether /heptane, followed by 1: 19 2M ammonia (in methanol)/ethyl acetate affords a brown foam (351 g). Crystallization of 320 g of this material from methyl t-butyl ether and heptane, affords the title compound (279.4 g) as an off-white solid after drying for 2 days at 45°C. LCMS (m/e): 560 (M+H); [αβ° = - 44°. Glucokinase Assay
The human islet GK isoform is expressed in E.coli as (His)6 -tagged fusion protein and purified with metal chelate affinity chromatography, see e.g. Tiedge et al., Biochem. Biophys. Acta 1337, 175-190, 1997. After purification the enzyme is stored in aliquots at concentration 0.8 mg/ml in 25 mM sodium phosphate, 150 mM sodium chloride, 100 mM imidazole, 1 mM dithiothreitol, 50 % glycerol at -800C. The assay is performed in flat bottom 96-well plates in a final incubation volume of lOOμL. The incubation mixture consists of 25 mM 4-(2-hydroxyethyl)-l-piperazineethanesulfonic acid (HEPES) (pH7.4), 50 mM potassiumchloride, 2.5 mM magnesiumchloride, 2 mM dithiothreitol, 4 U/ml glucose-6-phosphate dehydrogenase from Leuconostoc mesenteroides, 5 mM ATP, 1 mM NAD and a set concentration of glucose. Test compounds are dissolved in dimethylsulfoxide and then added to the reaction mixture giving the final dimethylsulfoxide concentration of 10%. The reaction is initiated by addition of 20 μL GK and run for 20 min at 37°C. The amount of formed NADH is measured as an increase in absorbance at 340 nm using a microplate reader. Absorbance values are used for EC50 calculations.
Example 1 activated GK with an EC50 = 42±42 nM (n=5) at 10 mM glucose. It also increased the enzyme activity in a concentration dependent manner at lower glucose concentrations.
Glycolysis Assay
Rat insulinoma INS-IE cells are maintained at 370C, 5% CO2, 95% humidity in 1640 medium supplemented with 11 mM glucose, 5% Fetal Bovine Serum, 50 μM 2- Mercaptoethanol, 1 mM pyruvate, 10 mM HEPES and antibiotics. Prior to assay, cells are trypsinized, pelleted by centrifugation and seeded into 96-well tissue culture assay plates at the density of 30,000 cells/well. Cells are allowed to attach and incubated for 48 hours at 370C, 5% CO2. On the assay day, cells are washed with and incubated in 200 μL Earle's Balanced Salt Solution (EBSS) buffer supplemented with 0.1% Bovine Serum Albumin (BSA). After 30 minutes incubation, the buffer is removed and 100 μL EBSS buffer containing 0.1% BSA, 8 mM glucose and the compound is added to cells. Immediately after, 20 μL of CellTiter 96® AQueous One Solution Reagent is added to cells and cells are incubated at 370C for an additional hour. At the end of incubation absorbance at 490 nm is read. Absorbance values are used for EC50 calculations.
Example 1 stimulates glucose metabolism in rat insulinoma INSl-E cells (mean EC50= 579±139 nM, n=4). Thus, the compounds of the present invention are shown to activate GK in vitro.
Oral Glucose Tolerance Test (OGTT)
Male Wistar rats at a weight of 225-250 g, are kept on regular diet and with normal light cycle and conditions. For the study, rats are fasted overnight before their exact weights are measured, and are randomized into groups of similar weights (n=4 per group). The compound is suspended in a 1 : 1 mixture of solutol/ethanol in a bath sonicator (10% of total volume). The obtained suspension is then diluted with 9 volumes of 10% aqueous solutol solution, and the compound is dosed orally at 1, 3, 6, 10, 20, and 30 mg/kg orally. Rats are given a 2 g/kg oral glucose bolus 2 hours after compound administration. Blood is collected via tail bleed at 0, 15, 30, 60, 90 and 120 minutes post glucose administration. Collected blood is placed into ethylenediaminetetraacetic acid (EDTA) tubes at volume of 400 μL per sample, and the samples are kept on ice. Plasma is isolated and stored at -2O0C until samples are analyzed for glucose and compound exposure. Area under the plasma glucose curve (glucose AUC) is calculated for each group and the percentage decrease in the glucose AUC versus the control group is used as a measure of efficacy of the compound to decrease plasma glucose.
Example 1 decreases plasma glucose in a dose-dependent manner at both fasted and postprandial glucose levels. A maximal lowering of glucose AUC versus the untreated control group is observed with the high dose (30 mg/kg) and represents a 42% decrease. Interpolation of the data showed that a 20% glucose AUC decrease occurs at an average compound concentration of 99 ng/ml (179 nM) in plasma, corresponding to a 6.9 mg/kg compound dose.
Thus, the compounds of the present invention are shown to activate GK in vivo.
Blood Brain Barrier Permeability
A stock compound solution is prepared in dimethylsulfoxide at 10 mM. A dose solution is then prepared at ImM by diluting 100 μL of the stock with 900 μL of propylene glycol. The dose is administered as an IV bolus (2.2 mL/kg) via the tail vein into six male CF-I mice (approximately 23 g) for a target dose of 2.17 μmole/kg. Mice are euthanized using both CO2 and cervical dislocation. Three mice are sacrificed 5 minutes post dose and three 60 minutes post dose. Blood is collected from each animal via cardiac puncture, and plasma is prepared using sodium EDTA, transferred into polypropylene sample tubes and immediately frozen using dry ice. Complete brain is collected from each animal and bissected medially, each half being transferred into polypropylene sample tubes and immediately frozen using dry ice. Plasma samples are prepared for analysis by precipitation of protein using two parts of extraction sovent (10% tetrahydrofuran in acetonitrile) to one part plasma and mixing with a vortex mixer. For brain tissue, it is assumed that 1 mg brain tissue ~ 1 μL volume and two parts of extraction solvent are added to one part tissue. The samples are immediately homogenized using an ultrasoinc cell dismemberator. Calibration standards are prepared by spiking known concentrations of compound into blank mouse plasma and then treated as plasma samples. All samples are centrifuged at 6000 RCF for 5 minutes. An aliquot of supernatant from each sample is transferred to a polypropylene 96 well plate and sealed for analysis by LC-MS/MS.
MS/MS is effected using a Sciex API 4000 triple quadrupole mass spectrometer equipped with a turbo ion spray source. High Performance Liquid Chromatogaphy is effected using a Phenomenex Hyrdro RP analytical column (10Ox 2.0 mm, 4 μ) heated to 500C and operated at a constant flow rate of 0.6 mL/minute. A mobile phase gradient is utilized consisting of an initial mobile phase of 60:40 5 mM aqueous ammonium formate:5mM ammonium formate in methanol with a hold time of 1 minute followed by a linear 2 minute gradient to 10:90 5 mM aqueous ammonium formate: 5 mM ammonium formate in methanol with a final hold time of 1 minute. Column effluent is diverted to waste from 0-2.8 minutes and then directed into the mass spectrometer from 2.8-4.0 minutes MS/MS transitions monitored are 560/84. Quantitation of compound in test samples is achieved by comparing peak area values to a quadratic equation weighted 1/x2 derived from the nominal concentrations of the calibration standards and their respective peak areas. Upper and Lower Limits of Quantitation are determined by the back calculated recoveries of calibration standards that exceeded +/- 20% of theory. Brain tissue concentrations are corrected for plasma contribution using a literature factor of 16 μL of plasma/gm mouse brain.
The in vivo blood brain barrier permeability of Example 1 resulted in a mean brain/plasma ratio of 0.17 five minutes post dose and a mean total brain level of 0.539 nmol/g at that time.
The compounds of the present invention have been shown to have limited blood brain barrier permeability and so provides limited potential for severe hypoglycaemia.

Claims

WE CLAIM:
1. A compound of the formula:
Figure imgf000013_0001
or a pharmaceutically acceptable salt thereof.
2. A compound according to claim 1 of the formula:
Figure imgf000013_0002
or a pharmaceutically acceptable salt thereof.
3. A pharmaceutical composition comprising a compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable diluent or carrier.
4. A compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, for use in therapy.
5. A compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, for use in the treatment of diabetes.
6. A compound according to claim 5, or a pharmaceutically acceptable salt thereof, for use in the treatment of type II diabetes.
7. A method for the treatment of diabetes, which comprises administering an effective amount of a compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, to a human being or animal in need thereof.
8. A method according to claim 7 for the treatment of type II diabetes.
9. A pharmaceutical composition for use in therapy comprising a compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof.
10. A pharmaceutical composition for use in the treatment of diabetes comprising a compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof.
11. A pharmaceutical composition according to claim 10 for use in the treatment of type II diabetes.
PCT/US2009/067603 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators WO2010080333A1 (en)

Priority Applications (21)

Application Number Priority Date Filing Date Title
UAA201107562A UA104742C2 (en) 2008-12-19 2009-11-12 Arylcyclopropylacetamide derivatives useful as glucokinase activators
NZ592930A NZ592930A (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators
BRPI0923183-8A BRPI0923183B1 (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glycokinase activators, and pharmaceutical composition
PL09768316T PL2379517T3 (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators
EA201170845A EA019055B1 (en) 2008-12-19 2009-12-11 Cyclopropyl derivative, pharmaceutical composition and method for treatment of diabetes
SI200930713T SI2379517T1 (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators
ES09768316T ES2435242T3 (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators
MX2011006434A MX2011006434A (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators.
SG2011044823A SG172256A1 (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators
CN200980150686XA CN102256952B (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators
EP09768316.3A EP2379517B1 (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators
JP2011542276A JP5497064B2 (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators
AU2009335931A AU2009335931B2 (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators
CA2746746A CA2746746C (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators
KR1020117014073A KR101290437B1 (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators
DK09768316.3T DK2379517T3 (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators
ZA2011/03944A ZA201103944B (en) 2008-12-19 2011-05-27 Arylcyclopropylacetamide derivatives useful as glucokinase activators
IL213210A IL213210A (en) 2008-12-19 2011-05-30 Arylcyclopropylacetamide derivatives useful as glucokinase activators
TN2011000294A TN2011000294A1 (en) 2008-12-19 2011-06-09 Arylcyclopropylacetamide derivatives useful as glucokinase activators
MA33945A MA32902B1 (en) 2008-12-19 2011-06-14 Arilcyclopropylacetamide derivatives are useful for the activation of glucokinase
HRP20130983AT HRP20130983T1 (en) 2008-12-19 2013-10-16 Arylcyclopropylacetamide derivatives useful as glucokinase activators

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP08380341 2008-12-19
EP08380341.1 2008-12-19
US15378109P 2009-02-19 2009-02-19
US61/153,781 2009-02-19

Publications (1)

Publication Number Publication Date
WO2010080333A1 true WO2010080333A1 (en) 2010-07-15

Family

ID=40545955

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/067603 WO2010080333A1 (en) 2008-12-19 2009-12-11 Arylcyclopropylacetamide derivatives useful as glucokinase activators

Country Status (34)

Country Link
US (1) US8063079B2 (en)
EP (1) EP2379517B1 (en)
JP (1) JP5497064B2 (en)
KR (1) KR101290437B1 (en)
CN (1) CN102256952B (en)
AR (1) AR074470A1 (en)
AU (1) AU2009335931B2 (en)
BR (1) BRPI0923183B1 (en)
CA (1) CA2746746C (en)
CO (1) CO6382120A2 (en)
CR (1) CR20110342A (en)
DK (1) DK2379517T3 (en)
DO (1) DOP2011000197A (en)
EA (1) EA019055B1 (en)
EC (1) ECSP11011150A (en)
ES (1) ES2435242T3 (en)
HR (1) HRP20130983T1 (en)
IL (1) IL213210A (en)
JO (1) JO2979B1 (en)
MA (1) MA32902B1 (en)
MX (1) MX2011006434A (en)
MY (1) MY180567A (en)
NZ (1) NZ592930A (en)
PA (1) PA8851901A1 (en)
PE (1) PE20120339A1 (en)
PL (1) PL2379517T3 (en)
PT (1) PT2379517E (en)
SG (1) SG172256A1 (en)
SI (1) SI2379517T1 (en)
TN (1) TN2011000294A1 (en)
TW (1) TWI395748B (en)
UA (1) UA104742C2 (en)
WO (1) WO2010080333A1 (en)
ZA (1) ZA201103944B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011123572A1 (en) 2010-03-31 2011-10-06 The Scripps Research Institute Reprogramming cells

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2931056C (en) * 2013-11-22 2023-02-28 The University Of Tokyo Carrier for use in delivering drug, conjugate, composition comprising same, and method for administrating same

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004063179A1 (en) 2003-01-06 2004-07-29 Eli Lilly And Company Substituted arylcyclopropylacetamides as glucokinase activators
WO2006058923A1 (en) 2004-12-03 2006-06-08 Novo Nordisk A/S Heteroaromatic glucokinase activators
WO2007051847A1 (en) 2005-11-03 2007-05-10 Prosidion Ltd Tricyclo substituted amides as glucokinase modulators

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2233660T3 (en) 2000-05-03 2005-06-16 F. Hoffmann-La Roche Ag GLUCOKINASA ALQUINIL FENIL HETEROAROMATIC ACTIVATORS.
MXPA02010795A (en) 2000-05-08 2003-03-27 Hoffmann La Roche Para amine substituted phenylamide glucokinase activators.
JP3839723B2 (en) 2000-05-08 2006-11-01 エフ.ホフマン−ラ ロシュ アーゲー Substituted phenylacetamide and its use as a glucokinase activator
MY141521A (en) * 2002-12-12 2010-05-14 Hoffmann La Roche 5-substituted-six-membered heteroaromatic glucokinase activators
PL378117A1 (en) 2003-02-11 2006-03-06 Prosidion Limited Tri(cyclo) substituted amide compounds
US7262196B2 (en) 2003-02-11 2007-08-28 Prosidion Limited Tri(cyclo) substituted amide glucokinase activator compounds
WO2004076420A1 (en) * 2003-02-26 2004-09-10 Banyu Pharmaceutical Co., Ltd. Heteroarylcarbamoylbenzene derivative
MXPA06012008A (en) 2004-04-21 2007-01-25 Prosidion Ltd Tri(cyclo) substituted amide compounds.
JP2008509898A (en) 2004-08-12 2008-04-03 プロシディオン・リミテッド Substituted phenylacetamides and their use as glucokinase activators
CA2613585A1 (en) 2005-07-01 2007-01-11 Novartis Ag Combination of a renin inhibitor and an insulin secretion enhancer or an insulin sensitizer
KR100965040B1 (en) 2005-07-11 2010-06-21 미쓰비시 타나베 파마 코퍼레이션 An oxime derivative and preparations thereof
WO2007017649A1 (en) * 2005-08-09 2007-02-15 Astrazeneca Ab Heteroarylcarbamoylbenzene derivatives for the treatment of diabetes
JP2009514835A (en) 2005-11-03 2009-04-09 プロシディオン・リミテッド Tricyclo-substituted amide
WO2007051846A1 (en) 2005-11-03 2007-05-10 Prosidion Ltd Tricyclo substituted amides
WO2008012227A2 (en) * 2006-07-24 2008-01-31 F. Hoffmann-La Roche Ag Pyrazoles as glucokinase activators
US8314247B2 (en) * 2007-01-10 2012-11-20 Mitsubishi Tanabe Pharma Corporation Hydrazone derivative
EP2275414B1 (en) 2008-04-28 2015-06-10 Kyorin Pharmaceutical Co., Ltd. Cyclopentylacrylamide derivative

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004063179A1 (en) 2003-01-06 2004-07-29 Eli Lilly And Company Substituted arylcyclopropylacetamides as glucokinase activators
WO2006058923A1 (en) 2004-12-03 2006-06-08 Novo Nordisk A/S Heteroaromatic glucokinase activators
WO2007051847A1 (en) 2005-11-03 2007-05-10 Prosidion Ltd Tricyclo substituted amides as glucokinase modulators

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011123572A1 (en) 2010-03-31 2011-10-06 The Scripps Research Institute Reprogramming cells
EP3199623A1 (en) 2010-03-31 2017-08-02 The Scripps Research Institute Reprogramming cells
EP3936608A1 (en) 2010-03-31 2022-01-12 The Scripps Research Institute Reprogramming cells

Also Published As

Publication number Publication date
EA201170845A1 (en) 2011-12-30
SG172256A1 (en) 2011-07-28
CN102256952B (en) 2013-12-11
MY180567A (en) 2020-12-02
BRPI0923183B1 (en) 2019-03-19
SI2379517T1 (en) 2013-10-30
PA8851901A1 (en) 2010-07-27
PL2379517T3 (en) 2014-01-31
JP5497064B2 (en) 2014-05-21
NZ592930A (en) 2013-01-25
BRPI0923183A2 (en) 2016-02-16
DOP2011000197A (en) 2011-09-15
KR101290437B1 (en) 2013-07-29
TN2011000294A1 (en) 2012-12-17
BRPI0923183A8 (en) 2017-09-12
EA019055B1 (en) 2013-12-30
CO6382120A2 (en) 2012-02-15
HRP20130983T1 (en) 2013-11-22
MX2011006434A (en) 2011-07-19
TW201029999A (en) 2010-08-16
UA104742C2 (en) 2014-03-11
DK2379517T3 (en) 2013-09-30
PT2379517E (en) 2013-11-21
EP2379517B1 (en) 2013-08-28
AR074470A1 (en) 2011-01-19
AU2009335931B2 (en) 2012-05-31
AU2009335931A1 (en) 2011-07-07
JO2979B1 (en) 2016-03-15
PE20120339A1 (en) 2012-04-16
ES2435242T3 (en) 2013-12-17
EP2379517A1 (en) 2011-10-26
CN102256952A (en) 2011-11-23
US20100160395A1 (en) 2010-06-24
TWI395748B (en) 2013-05-11
CA2746746A1 (en) 2010-07-15
US8063079B2 (en) 2011-11-22
CR20110342A (en) 2011-09-14
MA32902B1 (en) 2011-12-01
IL213210A (en) 2015-03-31
IL213210A0 (en) 2011-07-31
ECSP11011150A (en) 2011-07-29
KR20110086751A (en) 2011-07-29
JP2012512874A (en) 2012-06-07
CA2746746C (en) 2013-09-24
ZA201103944B (en) 2012-11-28

Similar Documents

Publication Publication Date Title
EP3010910B1 (en) Pyrimidinedione compounds against cardiac conditions
CA2904985C (en) Pyrazole-amide compound and medicinal uses therefor
US9855267B2 (en) Histone deacetylase inhibitors and compositions and methods of use thereof
KR20210034082A (en) 2,6-diamino pyridine compound
TW201643143A (en) Compound for inhibiting neuronal cell death induced by oxidative stress
AU2009335931B2 (en) Arylcyclopropylacetamide derivatives useful as glucokinase activators
US20050176813A1 (en) Novel chalcone derivatives and uses thereof
KR20140085580A (en) DRUGS FOR INHIBITING p38 AND USES THEREOF
EP4010324B1 (en) Pyrimidine-5-carboxamide compound
EA044754B1 (en) PYRIMIDINE-5-CARBOXAMIDE COMPOUND
EP2272844A1 (en) Five-membered ring compound
JP2012500807A (en) Octahydroquinolidine for antidiabetic treatment

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200980150686.X

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09768316

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 947/MUMNP/2011

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 592930

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 213210

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2009768316

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2009335931

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2746746

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 001220-2011

Country of ref document: PE

Ref document number: DZP2011000418

Country of ref document: DZ

WWE Wipo information: entry into national phase

Ref document number: MX/A/2011/006434

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2011542276

Country of ref document: JP

Kind code of ref document: A

Ref document number: 20117014073

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2011001483

Country of ref document: CL

Ref document number: 12011501248

Country of ref document: PH

Ref document number: CR2011-000342

Country of ref document: CR

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 11079303

Country of ref document: CO

ENP Entry into the national phase

Ref document number: 2009335931

Country of ref document: AU

Date of ref document: 20091211

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 201170845

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: a201107562

Country of ref document: UA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: PI0923183

Country of ref document: BR

ENP Entry into the national phase

Ref document number: PI0923183

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110620