WO2002046173A1 - Fused heteroaromatic glucokinase activators - Google Patents

Fused heteroaromatic glucokinase activators Download PDF

Info

Publication number
WO2002046173A1
WO2002046173A1 PCT/EP2001/013870 EP0113870W WO0246173A1 WO 2002046173 A1 WO2002046173 A1 WO 2002046173A1 EP 0113870 W EP0113870 W EP 0113870W WO 0246173 A1 WO0246173 A1 WO 0246173A1
Authority
WO
WIPO (PCT)
Prior art keywords
cyclopentyl
phenyl
propionamide
methanesulfonyl
mmol
Prior art date
Application number
PCT/EP2001/013870
Other languages
English (en)
French (fr)
Inventor
Wendy Lea Corbett
Joseph Samuel Grimbsy
Nancy-Ellen Haynes
Robert Francis Kester
Paige Erin Mahaney
Ramakanth Sarabu
Original Assignee
F. Hoffmann-La Roche Ag
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
Application filed by F. Hoffmann-La Roche Ag filed Critical F. Hoffmann-La Roche Ag
Priority to AU2190202A priority Critical patent/AU2190202A/xx
Priority to AU2002221902A priority patent/AU2002221902B2/en
Priority to JP2002547912A priority patent/JP4109111B2/ja
Priority to CA002429642A priority patent/CA2429642C/en
Priority to KR1020037007517A priority patent/KR100545431B1/ko
Priority to BR0115999-2A priority patent/BR0115999A/pt
Priority to EP01999565A priority patent/EP1341774B1/en
Priority to DE60117059T priority patent/DE60117059T2/de
Publication of WO2002046173A1 publication Critical patent/WO2002046173A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D215/38Nitrogen atoms
    • 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
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles 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 in position 2
    • C07D235/30Nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/52Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings condensed with carbocyclic rings or ring systems
    • C07D263/54Benzoxazoles; Hydrogenated benzoxazoles
    • C07D263/58Benzoxazoles; Hydrogenated benzoxazoles 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 in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • C07D277/68Benzothiazoles 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 in position 2
    • C07D277/82Nitrogen atoms

Definitions

  • Glucokinase is one of four hexokinases that are found in mammals [Colowick, S.P., in The Enzymes, Vol. 9 (P. Boyer, ed.) Academic Press, New York, NY, pages 1-48, 1973].
  • the hexokinases catalyze the first step in the metabolism of glucose, i.e., the conversion of glucose to glucose-6-phosphate.
  • Glucokinase has a limited cellular distribution, being found principally in pancreatic ⁇ -cells and liver parenchymal cells.
  • GK is a rate-controlling enzyme for glucose metabolism in these two cell types that are known to play critical roles in whole-body glucose homeostasis [Chipkin, S.R., Kelly, K.L., and Ruderman, N.B. in Joslin's Diabetes (CR. Khan and G.C. ier, eds.), Lea and Febiger, Philadelphia, PA, pages 97-115, 1994].
  • concentration of glucose at which GK demonstrates half-maximal activity is approximately 8 mM.
  • the other three hexokinases are saturated with glucose at much lower concentrations ( ⁇ 1 mM).
  • GK does indeed play a critical role in whole-body glucose homeostasis. Animals that do not express GK die within days of birth with severe diabetes while animals overexpressing GK have improved glucose tolerance (Grupe, A., Hultgren, B., Ryan, A. et al., Cell 83, 69-78, 1995; Feme, T., Riu, E., Bosch, F. et al., FASEB J., 10, 1213-1218, 1996). An increase in glucose exposure is coupled through GK in ⁇ -cells to increased insulin secretion and in hepatocytes to increased glycogen deposition and perhaps decreased glucose production.
  • This invention provides a compound, comprising an amide of the formula 1-0
  • this invention provides a compound, comprising an amide of the formulae la, lb, Ha or lib:
  • R 3 is a cycloalkyl having from 4 to 7 carbon atoms or 2-propyl;
  • R 5 is halogen, preferably Cl or F;
  • R 6 is halogen, preferably Cl or F;
  • R is an alkyl having from 1 to 3 carbon atoms; R is hydrogen, halo, nitro, cyano, or perfluoro-methyl; R 3 is a cycloalkyl having from 4 to 7 carbon atoms or 2- propyl; each Y is independently CH or N; dotted lines collectively represent 0 or 2 additional double bonds in the heterocyclic ring structure; or a pharmaceutically acceptable salt thereof; or
  • R 3 is a cycloalkyl having from 4 to 7 carbon atoms or 2-propyl; R is halogen, preferably Cl or F; R 6 is halogen, preferably Cl or F; each Y is independently CH or N; and dotted lines collectively represent 0 or 2 additional double bonds in the heterocyclic ring structure; or a pharmaceutically acceptable salt thereof.
  • the present invention also relates to a pharmaceutical composition comprising a compound of formula I-O and a pharmaceutically acceptable carrier and/or adjuvant. Furthermore, the present invention relates to the use of such compounds as therapeutic active substances as well as to their use for the preparation of medicaments for the treatment or prophylaxis of type II diabetes. The present invention further relates to processes for the preparation of the compounds of formula 1-0. In addition, the present invention relates to a method for the prophylactic or therapeutic treatment of type II diabetes, which method comprises administering a compound of formula I-O to a human being or an animal.
  • R 3 is a cyclopentyl group.
  • the dotted lines collectively represent zero or two, preferably two additional double bonds in the heterocyclic ring.
  • the dotted lines collectively represent zero or two, preferably two additional double bonds in the heterocyclic ring.
  • R 1 is CH 3 and R 2 is H.
  • examples of such amides are N-benzothiazol-2-yl-3-cyclopentyl-2-(4-methanesulfonyl- phenyl)-propionamide and 3-cyclopentyl-2-(4-methanesulfonyl-phenyl)-N-quinolin-2-yl- propionamide.
  • R 1 is SO 2 CH 3 and R 2 is halo.
  • examples of such amides are N-benzooxazol-2-yl-2(R)-(3-chloro-4- methanesulfonyl-phenyl)-3-cyclopentyl-propionamide; 2(R)-(3-chloro-4- methanesulfonyl-phenyl)-3-cyclopentyl-N-quinolin-2-yl-propionamide; N-(1H- benzoimidazol-2-yl)-2-(3-bromo-4-methanesulfonyl-phenyl)-3-cyclopentyl- propionamide; and 2-(3-bromo-4-methanesulfonyl-phenyl)-3-cyclopentyl-N-quinolin-2- yl-propionamide.
  • R 1 is CH 3 and R 2 is CN.
  • examples of such amides are N-benzothiazol-2-yl-2-(3-cyano-4-methanesulfonyl- phenyl)-3-cyclopentyl-propionamide; N-benzooxazol-2-yl-2-(3-cyano-4-methanesulfonyl- phenyl)-3-cyclopentyl-propionamide; N-(lH-benzoimidazol-2-yl)-2-(3-cyano-4- methanesulfonyl-phenyl)-3-cyclopentyl-propionamide; and 2-(3-cyano-4- methanesulfonyl-phenyl)-3-cyclopentyl-N-quinolin-2-yl-propionamide.
  • R 1 is CH 3 and R 2 is CF 3 .
  • amides are 3-cyclopentyl-2-(4-methanesulfonyl-3-trifluoromethyl- phenyl)-N-quinolin-2-yl-propionamide; N-benzothiazol-2-yl-3-cyclopentyl-2-(4- methanesulfonyl-3-trifluoromethyl-phenyl)-propionamide; N-(lH-benzoimidazol-2-yl)-3- cyclopentyl-2-(4-methanesulfonyl-3-trifluoromethyl-phenyl)-propionamide; and N- benzooxazol-2-yl-3-cyclopentyl-2-(4-methanesulfonyl-3-trifluoromethyl-phenyl)- propionamide.
  • amides of formulae la and Ea R 1 is CH 3 , and R 2 is NO 2 .
  • examples of such amides are N-benzothiazol-2-yl-3-cyclopentyl-2-(4-methanesulfonyl-3- nitro-phenyl)-propionamide; N-benzooxazol-2-yl-3-cyclopentyl-2-(4-methanesulfonyl-3- nitro-phenyl)-propionamide; N-(lH-benzoimidazol-2-yl)-3-cyclopentyl-2-(4- methanesulfonyl-3-nitro-phenyl)-propionamide; and 3-cyclopentyl-2-(4-methanesulfonyl- 3-nitro-phenyl)-N-quinolin-2-yl-propionamide.
  • W is O.
  • examples of such amides include N-benzooxazol-2-yl-3-cyclopentyl-2(R)-(3,4-dichloro-phenyl)-propionamide; and N-benzooxazol-2-yl-3-cyclopentyl-2-(4-methanesulfonyl-phenyl)-propionamide.
  • W is S.
  • amides examples include N-benzothiazol-2-yl-3-cyclopentyl-2-(3,4-dichloro-phenyl)-propionamide; N-benzothiazol-2-yl-2-(3-bromo-4-methanesulfonyl-phenyl)-3-cyclopentyl- propionamide; and N-benzothiazol-2-yl-2-(3-cyano-4-methanesulfonyl-phenyl)-3- cyclopentyl-propionamide.
  • amides of formulae la and lb W is NH.
  • examples of such amides include N-(lH-benzoimidazol-2-yl)-3-cyclopentyl-2-(3,4-dichloro-phenyl)- propionamide; and N-(lH-benzoimidazol-2-yl)-2-(3-chloro-4-methanesulfonyl-phenyl)-3- cyclopentyl-propionamide.
  • examples of such amides include 3-cyclopentyl-2- (3 ,4-dichloro-phenyl)-N-(6-fluoro-benzothiazol-2-yl)-propionamide; and 3-cyclopentyl-2- (3,4-dichIorophenyl)-N-(6-methanesulfonyl-benzothiazol-2-yl)-propionamide.
  • both R 5 and R 6 are Cl or both R 5 and R 6 are F. Most preferably, both R 5 and R 6 are Cl.
  • both Y are CH.
  • Examples of such amides include 3-cyclopentyl-2-(3,4-dichloro-phenyl)-N-quinolin-2-yl-propionamide; 3- cyclopentyl-2-(4-methanesulfonyl-phenyl)-N-quinolin-2-yl-propionamide; 2(R)-(3- chloro-4-methanesulfonyl-phenyl)-3-cyclopentyl-N-quinolin-2-yl-propionamide; 2-(3- chloro-4-methanesulfonyl-phenyl)-3-cyclopentyl-N-quinolin-2-yl-propionamide; 2-(3- bromo-4-methanesulfonyl-phenyl)-3-cyclopentyl-N-quinolin-2-yl-propionamide; 2-(3- cyano-4-me
  • amides of formulae Ea and Eb the dotted lines collectively represent two additional double bonds.
  • Examples of such amides include 3-cyclopentyl- 2-(3,4-dichloro-phenyl)-N-quinolin-2-yl-propionamide; 3-cyclopentyl-2-(4- methanesulfonyl-phenyl)-N-quinolin-2-yl-propionamide; 2(R)-(3-chloro-4- methanesulfonyl-phenyl)-3-cyclopentyl-N-quinolin-2-yl-propionamide; 2-(3-chloro-4- methanesulfonyl-phenyl)-3-cyclopentyl-N-quinolin-2-yl-propionamide; 2-(3-bromo-4- methanesulfonyl-phenyl)-3-cyclopentyl-N-quinolin-2-yl-propionamide; 2-(3-bromo
  • preferable substituent R 3 is a cycloalkyl having from 4 to 7 carbon atoms, with cyclopentyl being especially preferred.
  • preferable substituent R 1 is methyl.
  • preferable substituents R 5 and R 6 are chloro.
  • preferable substituent Y is CH and dotted lines preferably collectively represent 2 additional double bonds in the heterocyclic ring structure.
  • R 1 is methyl;
  • R 2 is hydrogen, halo, nitro, cyano, or perfluoro-methyl;
  • R 3 is cyclopentyl;
  • R 5 and R 6 are Cl;
  • W is O, S or NH;
  • each Y is CH;
  • R 1 is methyl
  • R is ccyyccllooppeennttyyll;
  • W is O, S or NH; and * denotes an asymmetric carbon.
  • R 1 is methyl;
  • R 2 is hydrogen, halo (preferably chloro or bromo), nitro, cyano, or perfluoro-methyl;
  • R 3 is cyclopentyl;
  • Y is CH; dotted lines collectively represent 2 additional double bonds in the heterocyclic ring structure; and * denotes an asymmetric carbon atom.
  • R 3 is cyclopentyl;
  • R 5 and R 6 are Cl;
  • Y is CH; dotted lines collectively represent 2 additional double bonds in the heterocyclic ring structure; and * denotes an asymmetric carbon atom.
  • Most preferable compounds in accordance with the present invention are N-Benzothiazol-2-yl-2(R)-(3-chloro-4-methanesulfonyl-phenyl)-3-cyclopentyl- propionamide, N-(lH-benzoimidazol-2-yl)-2-(3-chloro-4-methanesulfonyl-phenyl)-3-cyclopentyl- propionamide, and 2-(3-chloro-4-methanesulfonyl-phenyl)-3-cyclopentyl-N-quinolin-2-yl-propionamide.
  • halogen and the term "halo", unless otherwise stated, designate all four halogens, i.e. fluorine, chlorine, bromine and iodine.
  • a preferred halogen is chlorine.
  • lower alkyl includes both straight chain and branched chain alkyl groups having from 1 to 8 carbon atoms, preferably from 1 to 3 carbon atoms, such as methyl, ethyl, propyl, isopropyl, preferably methyl.
  • aryl signifies aryl mononuclear aromatic hydrocarbon groups such as phenyl, tolyl, etc.
  • aryl groups are the substituted and unsubstituted mononuclear aryl groups, particularly phenyl.
  • lower alkoxy includes both straight chain and branched chain alkoxy groups having from 1 to 7 carbon atoms, such as methoxy, ethoxy, propoxy, isopropoxy, preferably methoxy and ethoxy.
  • lower alkanoic acid denotes lower alkanoic acids containing from 2 to 7 carbon atoms such as propionic acid, acetic acid and the like.
  • aroyl denotes aroic acids wherein aryl is as defined hereinbefore, with the hydrogen group of the COOH moiety removed.
  • aryl is as defined hereinbefore, with the hydrogen group of the COOH moiety removed.
  • benzoyl is one preferred aroyl group.
  • lower alkyl thio means a lower alkyl group as defined above where a thio group is bound to the rest of the molecule.
  • lower alkyl sulfonyl means a lower alkyl group as defined above where a sulfonyl group is bound to the rest of the molecule.
  • cycloalkyl means a saturated hydrocarbon ring having from 3 to 10 carbon atoms, preferably from 3 to 7 carbon atoms.
  • a preferred cycloalkyl is cyclopentyl.
  • lower alkoxy includes both straight chain and branched chain alkoxy groups having from 1 to 7 carbon atoms, such as methoxy, ethoxy, propoxy, isopropoxy, preferably methoxy and ethoxy.
  • hydrolyzable ester or ether protecting groups designates any ester or ether conventionally used for protecting carboxylic acids or alcohols which can be hydrolyzed to yield the respective hydroxyl or carboxyl group.
  • ester groups useful for those purposes are those in which the acyl moieties are derived from a lower alkanoic, aryl lower alkanoic, or lower alkane dicarboxyclic acid.
  • activated acids which can be utilized to form such groups are acid anhydrides, acid halides, preferably acid chlorides or acid bromides derived from aryl or lower alkanoic acids.
  • Example of anhydrides are anhydrides derived from monocarboxylic acid such as acetic anhydride, benzoic acid anhydride, and lower alkane dicarboxcyclic acid anhydrides, e.g. succinic anhydride as well as chloro formates e.g. trichloro and ethylchloro formate being preferred.
  • a suitable ether protecting group for alcohols are, for example, the tetrahydropyranyl ethers such as 4-methoxy-5,6-dihydroxy- 2H-pyranyl ethers.
  • aroylmethylethers such as benzyl, benzhydryl or trityl ethers or ⁇ -lower alkoxy lower alkyl ethers, for example, methoxymethyl or allylic ethers or alkyl silylethers such as trimethylsilylether.
  • amino protecting group designates any conventional amino protecting group which can be cleaved to yield the free amino group.
  • the preferred protecting groups are the conventional amino protecting groups utilized in peptide synthesis. Especially preferred are those amino protecting groups which are cleavable under mildly acidic conditions from about pH 2.0 to 3. Particularly preferred amino protecting groups are t-butylcarbamate (BOC), benzylcarbamate (CBZ), and 9- fluorenylmethylcarbamate (FMOC).
  • pharmaceutically acceptable salts include any salt with both inorganic or organic pharmaceutically acceptable acids such as hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid, citric acid, formic acid, maleic acid, acetic acid, succinic acid, tartaric acid, methanesulfonic acid, p ⁇ r ⁇ -toluene sulfonic acid and the like.
  • pharmaceutically acceptable salts also includes any pharmaceutically acceptable base salt such as amine salts, trialkyl amine salts and the like. Such salts can be formed quite readily by those skilled in the art using standard techniques.
  • R ⁇ is Cl, F or an alkyl sulfone of 1 to 3 carbon atoms
  • R 12 is Cl or F when R 11 is Cl or F and R 12 is hydrogen, halo, nitro, cyano, or perfluoro-methyl when R 11 is an alkyl sulfone
  • R 3 , W, and Y are as above, the dotted lines represent 0 or 2 additional double bonds in the heterocyclic ring
  • R 14 is hydrogen when the dotted lines represent 0 additional double bonds
  • R 14 is hydrogen, halo, or an alkyl sulfone having from 1 to 3 carbon atoms when the dotted lines represent 2 additional double bonds
  • R 15 is a hydrolyzable ester group
  • X is a halogen atom, preferably Br or I.
  • the carboxylic acids of formula N are known wherein R 12 is hydrogen and R 11 is mercapto (4-mercaptophenylacetic acid), methylthio (4-methylthiophenylacetic acid), or methylsulfonyl (4-methylsulfonylphenylacetic acid).
  • the carboxylic acids of formula N wherein both of R ⁇ and R 12 are chloro or fluoro (3,4-dichlorophenylacetic acid and 3,4-difluorophenyl acetic acid, respectively) are known.
  • the carboxylic acid of formula N wherein R ⁇ is fluoro and R 12 is chloro is also known (3-chloro-4-fluorophenylacetic acid). necessary for further chemical modification to produce the desired substitutions at R ⁇ and R 12 , the carboxylic acids can be converted to the corresponding esters of lower alkyl alcohols using any conventional esterification methods.
  • E it is desired to produce compounds of formula N where R 12 is hydrogen and R 11 is lower alkyl sulfonyl the known 4-mercaptophenylacetic acid may be used as a starting material.
  • the compound of formula N where R 12 is hydrogen and R 11 is mercapto may be alkylated by conventional methods (for example, with an alkyl halide) to the corresponding lower alkyl thio compounds of formula N.
  • the lower alkyl thio compounds can then be converted to the corresponding lower alkyl sulfonyl compounds of formula N by oxidation. Any conventional method of oxidizing an alkyl thio substituent to the corresponding sulfone group can be utilized to effect this conversion.
  • R is trifluoromethyl and R is lower alkyl thio are available, they can be converted to the corresponding compounds of formula N where R 12 is trifluoromethyl and R 11 is lower alkyl sulfonyl using conventional oxidation procedures.
  • any conventional method of converting a primary carboxamide to a carboxylic acid or carboxylic ester can be used to effect this conversion (see for example, Greenlee, W. J.; Thorsett, E. D. /. Org. Chem., 1981, 5351). These compounds can then be converted to the compounds of formulae V or VI where R 12 is nitro and R 11 is lower alkyl thio. Any conventional method of nucleophilic displacement of an aromatic chlorine group with a lower alkyl thiol can be utilized to effect this conversion (see for example, Testaferri, L. et al.
  • any conventional method of nucleophilic displacement of an aromatic chlorine group with a lower alkane sulfinate can be utilized to effect this conversion (see for example, Ulman, A.; Urankar, E. /. Org. Chem., 1989, 4691).
  • the known 2-chlorothiophenol can be used as starting material.
  • the mercapto group may be alkylated by conventional methods (for example, with a lower alkyl halide) to the corresponding 2-chloro-l -lower alkyl thio benzenes. These compounds can then be converted to the corresponding 3- chloro-4-(lower alkyl thio)-phenyl acetic acids.
  • the 2-chloro-l -lower alkyl thio benzenes are acylated with a (lower alkyl)oxalyl chloride (such as methyloxalyl chloride or ethyloxalyl chloride) via a Friedel-Crafts acylation to produce the beta-keto carboxylic ester in the position para to the lower alkyl thio functional group.
  • the beta-keto carboxylic ester is next hydrolyzed by any conventional method to convert a beta-keto carboxylic ester to a beta-keto carboxylic acid.
  • the alkyl halide of formula VE is reacted with the compound of formula V to produce the compound of formula IX or reacted with the compound of formula VI to produce the compound of formula VEL
  • the compounds of formulae V and VI represent an organic acid and an organic acid derivative having an alpha carbon atom
  • the compound of formula VE is an alkyl halide so that alkylation occurs at the alpha carbon atom of this carboxylic acid. This reaction is carried out by any conventional means of alkylation of the alpha carbon atom of a carboxylic acid or a lower alkyl ester of a carboxylic acid.
  • an alkyl halide is reacted with the dianion of the acetic acid or the anion generated from an acetic acid ester.
  • the anion can be generated by using a strong organic base such as lithium diisopropylamide and n-butyl lithium as well as other organic lithium bases.
  • low boiling ether solvents are utilized such as tetrahydrofuran at low temperatures from -80°C to about -10°C being preferred. However any temperature from -80°C to room temperature can be used.
  • the compound of formula VEI can be converted to the compound of formula IX by any conventional procedure to convert a carboxylic acid ester to an acid.
  • the compound of formula IX is condensed with the compounds of formulae X or XI via conventional peptide coupling to produce the compounds of formulae I or E, respectively.
  • any conventional method of condensing a primary amine with a carboxylic acid can be utilized to effect this conversion.
  • the amine of formula X is a five-membered heteroaromatic ring fused with a aromatic ring which contains six ring members or fused with a saturated six-membered cycloalkyl ring.
  • the five-membered heteroaromatic ring contains 2 heteroatoms selected from the group consisting of oxygen, sulfur, or nitrogen and is connected by a ring carbon to the amine of the amide group shown in formula I.
  • This five-membered heteroaromatic ring contains a first nitrogen heteroatom adjacent to the connecting ring carbon atom, and the other heteroatoms defined by W can be sulfur, oxygen or nitrogen. There are no heteroatoms on the fusion points.
  • Such five-membered heteroaromatic fused rings defined by formula X include, for example, benzothiazole, benzoxazole, benzoimidazole, and tetrahydrobenzothiazole. These heteroaromatic rings are connected via a ring carbon atom to the amide group to form the amides of formula I.
  • the ring carbon atom of the heteroaromatic ring which is connected via the amide linkage to form the compound of formula I cannot contain any substituent.
  • the amine of formula XI is a six-membered heteroaromatic ring fused with a aromatic ring which contains six ring members or fused with a saturated six-membered cycloalkyl ring.
  • the six-membered heteroaromatic ring contains 1 to 3 nitrogen heteroatoms and is connected by a ring carbon to the amine of the amide group shown in formula E.
  • This six-membered heteroaromatic ring contains a first nitrogen heteroatom adjacent to the connecting ring carbon atom, and if present, Y defines the location of the other nitrogen heteroatoms. There are no heteroatoms on the fusion points.
  • Such six- membered heteroaromatic fused rings defined by formula XI include, for example, quinoline, quinazoline, quinoxaline, benzotriazine, and tetrahydroquinoline. These heteroaromatic rings are connected via a ring carbon atom to the amide group to form the amides of formula E. The ring carbon atom of the heteroaromatic ring which is connected via the amide linkage to form the compound of formula E cannot contain any substituent.
  • the compound of formulae I and E has an asymmetric carbon atom through which the group -CH 2 R 3 and the acid amide substituents are connected.
  • the preferred stereoconfiguration of this group is R.
  • R or the S isomer of the compounds of formulae I and E these compounds can be isolated as the desired isomer by any conventional chemical means.
  • the preferred chemical mean is the use of pseudoephredrine as a chiral auxiliary for the asymmetric alkylation of the phenylacetic acids of formula V (see for example, Myers, A.G. et al. J. Am. Chem. Soc. 1997, 6496).
  • the compounds of formula V where R 12 is lower alkyl thio and R 11 is as described above are first converted to the pseudoephedrine amides using lR,2R-(-)- pseudoephedrine as the desired enantiomer of pseudoephedrine.
  • Any conventional method of converting a carboxylic acid to a carboxamide can be utilized to effect this conversion.
  • the pseudoephedrine amides can undergo highly diastereoselective alkylations with alkyl halides to afford the ⁇ -substituted amide products corresponding to formula IX.
  • R carboxylic acids of formula TX where R 12 is lower alkyl thio and R 11 is as described above by conventional acidic hydrolysis methods to convert a carboxamide to a carboxylic acid.
  • R carboxylic acids of formula IX where R 12 is lower alkyl thio and R 11 is as described above can be converted to the R isomers of formulae I and E where R 12 is lower alkyl thio and R 11 is as described above.
  • any conventional method of condensing a primary amine with a carboxylic acid can be utilized to effect this conversion.
  • R carboxylic acids of formula IX where R 12 is lower alkyl thio and R n is as described above can first be oxidized to the R compounds of formula TX where R 12 is lower alkyl sulfonyl and R 1 is as described above. Any conventional method of oxidizing an alkyl thio substituent to the corresponding sulfone group can be utilized to effect this conversion. These compounds can be then converted 1 to the corresponding R compounds of formulae I and E where R is lower alkyl sulfonyl and R 11 is as described above. In carrying out this reaction, any conventional method of condensing a primary amine with a carboxylic acid, without racemization, can be utilized to effect this conversion.
  • Another chemical means to produce the R or S isomer of the compounds of formulae I or E is to react the compound of formula IX with an optically active base.
  • Any conventional optically active base can be utilized to carry out this resolution.
  • the preferred optically active bases are the optically active amine bases such as alpha- methylbenzylamine, quinine, dehydroabietylamine and alpha-methylnaphthylamine. Any of the conventional techniques utilized in resolving organic acids with optically active organic amine bases can be utilized in carrying out this reaction.
  • the compound of formula IX is reacted with the optically active base in an inert organic solvent medium to produce salts of the optically active amine with both the R and S isomers of the compound of formula IX.
  • temperatures and pressure are not critical and the salt formation can take place at room temperature and atmospheric pressure.
  • the R and S salts can be separated by any conventional method such as fractional crystallization. After crystallization, each of the salts can be converted to the respective compounds of formula IX in the R and S configuration by hydrolysis with an acid.
  • dilute aqueous acids i.e., from about 0.00 IN to 2N aqueous acids, such as aqueous sulfuric or aqueous hydrochloric acid.
  • aqueous acids such as aqueous sulfuric or aqueous hydrochloric acid.
  • the resolution of racemates of the compounds of the formula IX can also be achieved via the formation of corresponding diastereomeric esters or amides.
  • These diastereomeric esters or amides can be prepared by coupling the carboxylic acids of the formula IX with a chiral alcohol or a chiral amine. This reaction can be carried out using any conventional method of coupling a carboxylic acid with an alcohol or an amine.
  • the corresponding diastereomers of compounds of the formula TX can then be separated using any conventional separation methods.
  • the resulting pure diastereomeric esters or amides can then be hydrolyzed to yield the corresponding pure R or S isomers.
  • the hydrolysis reaction can be carried out using conventional known methods to hydrolyze an ester or an amide without racemization.
  • the separation of R and S isomers can also be achieved using an enzymatic ester hydrolysis of any lower alkyl esters corresponding to the compound of the formula IX (see for example, Ahmar, M.; Girard, C; Bioch, R, Tetrahedron Lett, 1989, 7053), which results in the formation of corresponding chiral acid and chiral ester.
  • the ester and the acid can be separated by any conventional method of separating an acid from an ester.
  • the configuration of formula IX which is produced by this method of resolution is carried out throughout the entire reaction scheme to produce the desired R or S isomers of formulae I and E.
  • All of the compounds of formulae la, lb, Ea and Eb which include the compounds set forth in the Examples, activate glucokinase in vitro by the procedure of Biological Activity Example A. In this manner, they increase the flux of glucose metabolism which causes increased insulin secretion. Therefore, the compounds of formulae la, lb, Ea and Eb are glucokinase activators useful for increasing insulin secretion.
  • medicaments containing a compound of formula I-O are also an object of the present invention, as is a process for the manufacture of such medicaments, which process comprises bringing one or more compounds of formula I-O and, if desired, one or more other therapeutically valuable substances into a galenical administration form, e.g. by combining a compound of formula I-O with a pharmaceutically acceptable carrier and/or adjuvant.
  • compositions may be administered orally, for example in the form of tablets, coated tablets, dragees, hard or soft gelatine capsules, solutions, emulsions or suspensions.
  • Administration can also be carried out rectally, for example using suppositories; locally or percutaneously, for example using ointments, creams, gels or solutions; or parenterally, e.g. intravenously, intramuscularly, subcutaneously, intrathecally or transdermally, using for example injectable solutions.
  • administration can be carried out sublingually or as an aerosol, for example in the form of a spray.
  • the compounds of the present invention may be admixed with pharmaceutically inert, inorganic or organic excipients.
  • suitable excipients for tablets, dragees or hard gelatine capsules include lactose, maize starch or derivatives thereof, talc or stearic acid or salts thereof.
  • suitable excipients for use with soft gelatine capsules include for example vegetable oils, waxes, fats, semi-solid or liquid polyols etc.; according to the nature of the active ingredients it may however be the case that no excipient is needed at all for soft gelatine capsules.
  • excipients which may be used include for example water, polyols, saccharose, invert sugar and glucose.
  • excipients which may be used include for example water, alcohols, polyols, glycerine, and vegetable oils.
  • excipients which may be used include for example natural or hardened oils, waxes, fats and semi-solid or liquid polyols.
  • the pharmaceutical compositions may also contain preserving agents, solubilising agents, stabilising agents, wetting agents, emulsifiers, sweeteners, colorants, odorants, salts for the variation of osmotic pressure, buffers, coating agents or antioxidants. As mentioned earlier, they may also contain other therapeutically valuable agents. It is a prerequisite that all adjuvants used in the manufacture of the preparations are non-toxic.
  • Preferred forms of use are intravenous, intramuscular or oral administration, most preferred is oral administration.
  • the dosages in which the compounds of formula I-O are administered in effective amounts depend on the nature of the specific active ingredient, the age and the requirements of the patient and the mode of application. In general, dosages of about 1-100 mg/kg body weight per day come into consideration.
  • the bright yellow solution was allowed to stir at -78°C for 1 h, at which time, a solution of iodomethylcyclopentane (11.17 g, 53.2 mmol) in hexamethylphosphoramide (10 mL) was added via a cannula.
  • the reaction mixture was stirred at -78°C for 1 h.
  • the reaction mixture was then allowed to warm to 25°C where it was stirred for 14 h.
  • reaction mixture was stirred at 0°C for 30 min and then treated with a solution of 2-aminobenzothiazole (0.087 g, 0.58 mmol) in methylene chloride (2.5 mL) followed by NN-diisopropylethylamine (0.2 mL, 2.14 mmol).
  • the reaction mixture was stirred at 25°C for 16 h.
  • the reaction mixture was then diluted with water (10 mL) and extracted with methylene chloride (3 x 10 mL).
  • reaction mixture was then recooled to -78°C.
  • the second reaction mixture was added to the first reaction mixture over a period of 5 min via cannula.
  • the combined reaction was then stirred at -78°C for 15 min and then allowed to warm to 25°C where it was stirred for an additional 1.5 h.
  • the reaction was quenched by the addition of a saturated aqueous sodium bisulfite solution (50 mL) and extracted with ethyl acetate (3 x 40 mL).
  • reaction was quenched with a saturated aqueous sodium sulfite solution (16 mL) followed by the addition of a 0.5N aqueous sodium bicarbonate solution (50 mL).
  • a 0.5N aqueous sodium bicarbonate solution 50 mL.
  • the tetrahydrofuran was then removed in vacuo.
  • the residue was diluted with water (40 mL) and extracted with methylene chloride (3 x 20 mL).
  • methylene chloride (3 x 20 mL).
  • reaction mixture was then treated with 2- aminobenzothiazole (157 mg, 1.04 mmol) and pyridine (0.17 mL, 2.09 mmol), and the reaction mixture was allowed to stir at 25°C for 20 h.
  • the resulting reaction mixture was diluted with water (15 mL) and then extracted with methylene chloride (3 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then treated with a solution of 6-fluoro-benzothiazol-2-ylamine (123 mg, 0.73 mmol) in tetrahydrofuran (1.7 mL) and N,N-diisopropylethylamine (0.14 mL, 0.79 mmol). This solution was stirred at 25°C for l8 h. At this time, the reaction was concentrated in vacuo.
  • the reaction mixture was then diluted with water (10 mL) and extracted with methylene chloride (3 x 10 mL).
  • the combined organic layers were sequentially washed with water (1 x 10 mL), a IN aqueous sodium hydroxide solution (1 x 10 mL), a IN aqueous hydrochloric acid solution (1 x 10 mL), and a saturated aqueous sodium chloride solution (1 x 10 mL).
  • the organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then treated with 2-aminobenzoxazole (140 mg, 1.04 mmol) and pyridine (0.17 mL, 2.09 mmol), and the reaction mixture was allowed to stir at 25°C for 20 h.
  • the resulting reaction mixture was diluted with water (15 mL) and then extracted with methylene chloride (3 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then treated with 2-aminobenzimidazole (139 mg, 1.04 mmol) and pyridine (0.17 mL, 2.09 mmol), and the reaction mixture was allowed to stir at 25°C for 20 h.
  • the resulting reaction mixture was diluted with water (15 mL) and then extracted with methylene chloride (3 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • the resulting reaction mixture was then treated with a solution of 2-aminoquinoline (75 mg, 0.52 mmol) and pyridine (0.14 mL, 1J4 mmol) in tetrahydrofuran (5 mL), and the reaction mixture was allowed to warm to 25°C. The reaction was then stirred at 25°C for 16 hours. The reaction mixture was diluted with water (10 mL) and extracted with methylene chloride (3 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • the resulting reaction mixture was stirred at -78°C for 30 min and then treated dropwise with a solution of 4-(methylthio)phenylacetic acid (2.01 g, 11.03 mmol) in dry tetrahydrofuran (10.3 mL) and l,3-dimethyl-3,4,5,6-tetrahydro- 2(lH)-pyrimidinone (3.4 mL).
  • the reaction mixture was allowed to stir at -78°C for 1 h, at which time, a solution of iodomethylcyclopentane (2.55 g, 12.13 mmol) in a small amount of dry tetrahydrofuran was added dropwise.
  • reaction mixture was stirred at -78°C for 30 min and then allowed to warm to 25°C where it was stirred for 24 h.
  • the reaction mixture was quenched with water and then concentrated in vacuo to remove tetrahydrofuran.
  • the organic layer was washed with a saturated aqueous sodium chloride solution (1 x 100 mL), dried over sodium sulfate, filtered, and concentrated in vacuo.
  • the organic layer was sequentially washed with a IN aqueous hydrochloric acid solution (1 x 10 mL), water (1 x 10 mL), and a saturated aqueous sodium chloride solution (1 x 10 mL). The organic layer was dried over magnesium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was stirred at -10°C to -5°C for 30 min and then treated with lR,2R-(-)-pseudoephedrine (59.5 g, 353 mmol) in portions over 15 min while maintaining the temperature of the reaction mixture between -10°C and -4°C.
  • the reaction mixture was then stirred at -7°C to 0°C for 3 h.
  • the reaction mixture was then quenched at 0°C by the addition of water (150 mL). After vigorously stirring for 10 min, toluene (150 mL) was added, and the reaction mixture was stirred for 5 min. The organic layer was separated and washed with water (2 x 100 mL).
  • the combined aqueous layers were back-extracted with toluene (1 x 50 mL).
  • the combined organic layers were washed with a IN aqueous sulfuric acid solution (1 x 200 mL), a saturated aqueous sodium bicarbonate solution (1 x 200 mL), and a solution of water/saturated aqueous sodium chloride solution (1:1, 1 x 50 mL).
  • the resulting organic layer was then concentrated in vacuo to afford a white solid.
  • reaction mixture was stirred at -20°C for 30 min and then was treated with a solution of N-[2(R)-hydroxy-l(R)-methyl-2(R)-phenyl- ethyl]-N-methyl-2-(4-methylsulfanyl-phenyl)-acetamide (66.1 g, 201 mmol) in tetrahydrofuran (500 mL) over 50 min while maintaining the temperature between -20°C and -15°C.
  • the resulting yellow solution was stirred at 0°C for 30 min and then treated with a premixed solution of l,3-dimethyl-3,4,5,6-tetrahydro-2(lH)-pyrimidinone (51 mL, 418 mmol) and iodomethylcyclopentane (50.6 g, 239 mmol) over 30 min.
  • the resulting reaction mixture was stirred at 0°C for 4 h, at which time, thin layer chromatography analysis indicated that the reaction was complete.
  • the reaction mixture was then poured into toluene (400 mL).
  • the organic phase was washed sequentially with a solution of water/saturated aqueous sodium chloride solution (1:1, 1 x 1000 mL), a solution of water/saturated aqueous sodium chloride solution (1:2, 1 x 1000 mL), a 1M aqueous sulfuric acid solution (1 x 800 mL), water (1 x 200 mL), and a saturated aqueous sodium bicarbonate solution (1 x 1000 mL).
  • the silica gel plug was then washed with a solution of hexanes/ethyl acetate (4:1, 1.5 L), and the combined eluates were concentrated in vacuo.
  • the resulting pale-yellow oil was dissolved in ethyl acetate (35 mL) and subsequently treated with hexanes (100 mL).
  • the solution was stored in a refrigerator overnight.
  • the resulting solid was collected by filtration, washed with cold hexanes (ca.
  • the resulting reaction mixture was stirred at 25 °C for 19 h.
  • the reaction mixture was then concentrated in vacuo to remove methylene chloride.
  • the remaining black residue was diluted with a 10% aqueous hydrochloric acid solution (40 mL) and then extracted with ethyl acetate (3 x 25 mL).
  • the combined organic layers were washed with a saturated aqueous sodium chloride solution (1 x 20 mL), dried over sodium sulfate, filtered, and concentrated in vacuo.
  • the resulting reaction mixture was stirred at 25°C for 19 h.
  • the reaction mixture was then concentrated in vacuo to remove methylene chloride.
  • the remaining black residue was diluted with a 10% aqueous hydrochloric acid solution (40 mL) and then extracted with ethyl acetate (3 x 25 mL).
  • the combined organic layers were washed with a saturated aqueous sodium chloride solution (1 x 20 mL), dried over sodium sulfate, filtered, and concentrated in vacuo.
  • the reaction was then stirred at 25°C until the reaction temperature cooled back to 80°C. At this time, another portion of potassium hydroxide (2.09 g, 31.7 mmol) was added. Again, an exotherm was observed, and the resulting reaction mixture was allowed to cool back to 80°C. Once at 80°C, a third portion of potassium hydroxide (2.09 g, 31.7 mmol) was added to the reaction mixture. Another exotherm was observed, and after cooling back to 80°C, the fourth and final portion of potassium hydroxide (2.09 g, 31.7 mmol) was added. At this point, the heating element was added, and the reaction mixture was heated at 100°C for 16 h.
  • the resulting homogenous reaction mixture was cooled to 25°C and then diluted with water (12 mL). The reaction mixture was then transferred to a separatory funnel, rinsing with additional water (12 mL) and diethyl ether (40 mL). The layers were separated, and the aqueous layer was transferred to a flask. The organic layer was extracted with water (2 x 15 mL) The aqueous layers were combined and treated with heptane (20 mL), and the resulting reaction mixture was vigorously stirred. This stirred solution was then treated dropwise with concentrated hydrochloric acid (26 mL) over 30 min while the temperature was kept under 50°C with an ice bath.
  • reaction mixture was then treated with 2- aminobenzoxazole (121 mg, 0.91 mmol) and pyridine (0.15 mL, 1.8 mmol) and was stirred at 25°C for 16 h.
  • the reaction was then diluted with water (15 mL) and then extracted with methylene chloride (3 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then treated with 2-aminobenzoxazole (121 mg, 0.91 mmol) and pyridine (0.15 mL, 1.8 mmol), and the resulting reaction mixture was stirred at 25°C for 16 h.
  • the reaction was then diluted with water (15 mL) and extracted with methylene chloride (3 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then treated with 2-aminobenzothiazole (136 mg, 0.91 mmol) and pyridine (0.15 mL, 1.8 mmol) and was stirred at 25°C for 16 h.
  • the reaction was then diluted with water (15 mL) and then extracted with methylene chloride (3 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture became yellow in color, and a precipitate formed.
  • the reaction mixture was then heated at 80°C for 36 h.
  • the reaction mixture was allowed to cool to 25°C and then concentrated in vacuo to remove toluene.
  • the resulting residue was diluted with ethyl acetate (150 mL).
  • the organic layer was washed with a IN aqueous hydrochloric solution (1 x 100 mL), a 10% aqueous sodium carbonate solution (1 x 100 mL), and a saturated aqueous sodium chloride solution (1 x 100 mL).
  • the organic layer was then dried over magnesium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was quenched with a 1.5N aqueous sodium sulfite solution (25 mL) and then diluted with water (150 mL).
  • the aqueous layer was extracted with diethyl ether (3 x 50 mL).
  • the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then treated with 2-aminobenzothiazole (136 mg, 0.91 mmol) and pyridine (0.15 mL, 1.81 mmol), and the reaction mixture was stirred at 25°C for 16 h.
  • the reaction was then diluted with water (15 mL) and then extracted with methylene chloride (3 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then treated with 2-aminobenzimidazole (60 mg, 0.45 mmol) and pyridine (0.073 mL, 0.91 mmol). The resulting reaction mixture was stirred at 25°C for 16 h. The reaction was then diluted with water (15 mL) and then extracted with methylene chloride (3 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then treated with 2-aminobenzimidazole (121 mg, 0.91 mmol) and pyridine (0.15 mL, 1.82 mmol), and the resulting reaction mixture was stirred at 25°C for 16 h.
  • the reaction mixture was then diluted with water (15 mL) and extracted with methylene chloride (3 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then treated with a solution of 2-aminoquinoline (33 mg, 0.23 mmol) and pyridine (0.06 mL, 0J55 mmol) in NN-dimethylformamide (2.5 mL).
  • the resulting reaction mixture was allowed to warm to 25°C where it was stirred for 16 h.
  • the reaction mixture was then diluted with water (10 mL) and extracted with ethyl acetate (3 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then treated with a solution of 2-aminoquinoline (131 mg, 0.91 mmol) and pyridine (0.25 mL, 3.03 mmol) in NN-dimethylformamide (10 mL).
  • the resulting reaction mixture was allowed to warm to 25°C where it was stirred for 16 h.
  • the reaction mixture was then diluted with water (10 mL) and extracted with ethyl acetate (3 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • the resulting reaction mixture was stirred at -78°C for 30 min and then treated dropwise with a solution of (3-bromo-4- methylsulfanyl-phenyl)-acetic acid methyl ester (8.57 g, 31.15 mmol) in dry tetrahydrofuran (30 mL) and l,3-dimethyl-3,4,5,6-tetrahydro-2(lH)-pyrimidinone (10 mL).
  • the resulting reaction mixture was allowed to stir at -78°C for 1 h, at which time, a solution of iodomethylcyclopentane (7.85 g, 37.38 mmol) in a small amount of dry tetrahydrofuran was added dropwise.
  • reaction mixture was allowed to warm to 25°C where it was stirred for 15 h.
  • the reaction mixture was quenched with water (300 mL) and then concentrated in vacuo to remove tetrahydrofuran.
  • the remaining aqueous phase was extracted with ethyl acetate (3 x 150 mL).
  • the combined organic layers were washed with a saturated aqueous sodium chloride solution (1 x 200 mL), dried over sodium sulfate, filtered, and concentrated in vacuo.
  • the organic layer was washed sequentially with a IN aqueous hydrochloric acid solution (1 x 10 mL), water (1 x 10 mL), and a saturated aqueous sodium bicarbonate solution (1 x 10 mL). The organic layer was then dried over magnesium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then treated with NN- diisopropylethylamine (0.11 mL, 0.63 mmol) followed by a solution of 2-aminoquinoline (92 mg, 0.56 mmol) in dry tetrahydrofuran (3 mL). The resulting reaction mixture was stirred at 25°C for 17 h. The reaction mixture was concentrated in vacuo.
  • reaction mixture was stirred at -78°C for 30 min and then treated dropwise with a solution of (3-bromo-4-methylsulfanyl- phenyl)-acetic acid methyl ester (6.10 g, 22.17 mmol) in dry tetrahydrofuran (21 mL) and l,3-dimethyl-3,4,5,6-tetrahydro-2(lH)-pyrimidinone (7 mL). The resulting reaction mixture was allowed to stir at -78°C for 1 h, at which time, a solution of iodomethylcyclopentane (5.59 g, 26.60 mmol) in a small amount of dry tetrahydrofuran was added dropwise.
  • reaction mixture was allowed to warm to 25°C where it was stirred for 15 h.
  • the reaction mixture was quenched with water (300 mL) and then concentrated in vacuo to remove tetrahydrofuran.
  • the remaining aqueous phase was extracted with ethyl acetate (3 x 150 mL).
  • the combined organic layers were washed with a saturated aqueous sodium chloride solution (1 x 200 mL), dried over sodium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then diluted with water (40 mL), a IN aqueous hydrochloric acid solution (5 mL), and ethyl acetate (40 mL). The layers were separated, and the organic layer was dried over magnesium sulfate, filtered, and concentrated in vacuo.
  • the resulting yellow gel was diluted with methylene chloride (2 mL) and then slowly added to a solution of 2-aminoquinoline (84 mg, 0.583 mmol) and triethylamine (0.108 mL, 0.778 mmol) in NN- dimethylformamide (2 mL). The resulting reaction mixture was stirred at 25°C for 20 h. The reaction mixture was then diluted with water (25 mL), a IN aqueous hydrochloric acid solution (5 mL), and ethyl acetate (25 mL). The layers were separated, and the organic layer was dried over magnesium sulfate, filtered, and concentrated in vacuo.
  • reaction was treated with a solution of iodomethylcyclopentane (1.16 g, 5.52 mmol) in l,3-dimethyl-3,4,5,6-tetrahydro-2(lH)- pyrimidinone (1.2 mL).
  • the reaction mixture was stirred at -78°C for 4 h.
  • the reaction was then warmed to 25°C and was stirred at 25°C for 48 h.
  • This solution was then quenched by the slow addition of the reaction mixture to a 2N aqueous hydrochloric acid solution (50 mL).
  • the product was extracted into ethyl acetate (3 x 100 mL) and diethyl ether (1 x 50 mL).
  • the solution was sequentially washed with a saturated aqueous sodium bisulfite solution (2 x 50 mL), water (1 x 50 mL), a saturated aqueous sodium chloride solution (3 x 75 mL), a saturated aqueous sodium bicarbonate solution (1 x 75 mL), and a saturated aqueous sodium chloride solution (3 x 75 mL).
  • reaction mixture was then treated with a solution of 2-aminoquinoline (153 mg, 1.06 mmol) in tetrahydrofuran (2 mL) and triethylamine (0.17 mL, 1.20 mmol). This solution was stirred at 25°C for 50 h. At this time, the reaction was concentrated in vacuo.
  • reaction mixture was then diluted with methylene chloride (25 mL) and washed with a 3N aqueous hydrochloric acid solution (1 x 25 mL), water (1 x 25 mL), and a saturated aqueous sodium chloride solution (3 x 25 mL).
  • methylene chloride 25 mL
  • 3N aqueous hydrochloric acid solution 1 x 25 mL
  • water 1 x 25 mL
  • saturated aqueous sodium chloride solution 3 x 25 mL
  • the pale yellow reaction mixture was stirred at -78°C for 20 min and then slowly treated with a solution of 4-chloro-3-nitrophenylacetic acid methyl ester (5.00 g, 21.8 mmol) in a small amount of tetrahydrofuran over a 15 min period.
  • the reaction mixture turned deep purple (almost black) in color.
  • the reaction mixture was then stirred at -78°C for 1 h, at which time, a solution of iodomethylcyclopentane (4.58 g, 21.8 mol) in a small amount of dry tetrahydrofuran was added dropwise.
  • the reaction mixture was then stirred at -78°C and then allowed to warm to 25°C where it was stirred for 48 h.
  • reaction mixture was quenched with a saturated aqueous ammonium chloride solution (50 mL), and the resulting reaction mixture was concentrated in vacuo to remove tetrahydrofuran. The remaining residue was diluted with ethyl acetate (150 mL) and water (50 mL). The organic phase was washed with a saturated aqueous sodium chloride solution, dried over magnesium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then diluted with water (25 mL), a IN aqueous hydrochloric acid solution (5 mL), and ethyl acetate (25 mL). The layers were separated. The resulting organic layer was washed with a saturated aqueous sodium chloride solution (1 x 25 mL), dried over magnesium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then diluted with water (25 mL), a IN aqueous hydrochloric acid solution (5 mL), and ethyl acetate (25 mL). The layers were separated. The resulting organic layer was washed with a saturated aqueous sodium chloride solution (1 x 25 mL), dried over magnesium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was then diluted with water (25 mL), a IN aqueous hydrochloric acid solution (5 mL), and ethyl acetate (25 mL). The layers were separated. The resulting organic layer was washed with a saturated aqueous sodium chloride solution (1 x 25 mL), dried over magnesium sulfate, filtered, and concentrated in vacuo.
  • the assay was conducted at 25° C in a flat bottom 96-well tissue culture plate from Costar (Cambridge, MA) with a final incubation volume of 120 ⁇ L.
  • the incubation mixture contained: 25 mM Hepes buffer (pH, 7.1), 25 mM KCl, 5 mM D- glucose, ImM ATP, 1.8 mM NAD, 2 mM MgCl 2 , 1 ⁇ M sorbitol-6-phosphate, 1 mM dithiothreitol, test drug or 10% DMSO, 1.8 unit/ml G6PDH, and GK (see below).
  • C57BL/6J mice are orally dosed via gavage with Glucokinase (GK) activator at 50 mg/kg body weight following a two hour fasting period. Blood glucose - determinations are made five times during the six hour post-dose study period.
  • GK Glucokinase
  • GK activators are formulated at 6.76 mg/ml in Gelucire vehicle (Ethanol:Gelucire44/14:PEG400q.s. 4:66:30 v/w/v.
  • Mice are dosed orally with 7.5 ⁇ L formulation per gram of body weight to equal a 50 mg/kg dose.
  • a pre dose (time zero) blood glucose reading is acquired by snipping off a small portion of the animals tail ( ⁇ lmm) and collecting 15 ⁇ L blood into a heparinized capillary tube for analysis.
  • Tablets containing the following ingredients can be produced in a conventional manner:
  • Capsules containing the following ingredients can be produced in a conventional manner:
PCT/EP2001/013870 2000-12-06 2001-11-28 Fused heteroaromatic glucokinase activators WO2002046173A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
AU2190202A AU2190202A (en) 2000-12-06 2001-11-28 Fused heteroaromatic glucokinase activators
AU2002221902A AU2002221902B2 (en) 2000-12-06 2001-11-28 Fused heteroaromatic glucokinase activators
JP2002547912A JP4109111B2 (ja) 2000-12-06 2001-11-28 縮合複素環式芳香族グルコキナーゼアクチベーター
CA002429642A CA2429642C (en) 2000-12-06 2001-11-28 Fused heteroaromatic glucokinase activators
KR1020037007517A KR100545431B1 (ko) 2000-12-06 2001-11-28 축합된 이종방향족 글루코키나제 활성화제
BR0115999-2A BR0115999A (pt) 2000-12-06 2001-11-28 Composto, composição farmacêutica que compreende o mesmo, sua utilização, processo para o tratamento profilático ou terapêutico de diabetes do tipo ii e processo para a preparação do composto
EP01999565A EP1341774B1 (en) 2000-12-06 2001-11-28 Fused heteroaromatic glucokinase activators
DE60117059T DE60117059T2 (de) 2000-12-06 2001-11-28 Kondensierte heteroaromatische glucokinaseaktivatoren

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US25163700P 2000-12-06 2000-12-06
US60/251,637 2000-12-06

Publications (1)

Publication Number Publication Date
WO2002046173A1 true WO2002046173A1 (en) 2002-06-13

Family

ID=22952803

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2001/013870 WO2002046173A1 (en) 2000-12-06 2001-11-28 Fused heteroaromatic glucokinase activators

Country Status (21)

Country Link
US (3) US6545155B2 (US06448399-20020910-C00018.png)
EP (1) EP1341774B1 (US06448399-20020910-C00018.png)
JP (1) JP4109111B2 (US06448399-20020910-C00018.png)
KR (1) KR100545431B1 (US06448399-20020910-C00018.png)
CN (1) CN1226294C (US06448399-20020910-C00018.png)
AR (1) AR031627A1 (US06448399-20020910-C00018.png)
AT (1) ATE316965T1 (US06448399-20020910-C00018.png)
AU (2) AU2002221902B2 (US06448399-20020910-C00018.png)
BR (1) BR0115999A (US06448399-20020910-C00018.png)
CA (1) CA2429642C (US06448399-20020910-C00018.png)
DE (1) DE60117059T2 (US06448399-20020910-C00018.png)
DK (1) DK1341774T3 (US06448399-20020910-C00018.png)
ES (1) ES2256340T3 (US06448399-20020910-C00018.png)
GT (1) GT200100243A (US06448399-20020910-C00018.png)
MX (1) MXPA03005119A (US06448399-20020910-C00018.png)
PA (1) PA8534301A1 (US06448399-20020910-C00018.png)
PE (1) PE20020753A1 (US06448399-20020910-C00018.png)
PT (1) PT1341774E (US06448399-20020910-C00018.png)
UY (1) UY27055A1 (US06448399-20020910-C00018.png)
WO (1) WO2002046173A1 (US06448399-20020910-C00018.png)
ZA (1) ZA200303748B (US06448399-20020910-C00018.png)

Cited By (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004002481A1 (en) * 2002-06-27 2004-01-08 Novo Nordisk A/S Aryl carbonyl derivatives as therapeutic agents
WO2004045614A1 (en) * 2002-11-19 2004-06-03 Astrazeneca Ab Quinoline derivatives as glucokinase ligands
WO2004050645A1 (en) * 2002-10-03 2004-06-17 Novartis Ag Substituted (thiazol-2-yl) -amide or sulfonamide as glycokinase activators useful in the treatment of type 2 diabetes
WO2005095418A1 (en) * 2004-04-02 2005-10-13 Novartis Ag Sulfonamide-thiazolpyridine derivatives as glucokinase activators useful the treatment of type 2 diabetes
WO2005095417A1 (en) * 2004-04-02 2005-10-13 Novartis Ag Thiazolopyridine derivates, pharmaceutical conditions containing them and methods of treating glucokinase mediated conditions
WO2005103021A1 (en) * 2004-04-21 2005-11-03 Prosidion Limited Tri(cyclo) substituted amide compounds
JP2006517590A (ja) * 2003-02-11 2006-07-27 プロシディオン・リミテッド トリ(シクロ)置換されたアミド化合物
US7199140B2 (en) 2001-06-26 2007-04-03 Astrazeneca Ab Vinyl phenyl derivatives as GLK activators
WO2007039177A2 (en) 2005-09-29 2007-04-12 Sanofi-Aventis Phenyl- and pyridinyl- 1, 2 , 4 - oxadiazolone derivatives, processes for their preparation and their use as pharmaceuticals
WO2007096034A1 (en) * 2006-02-22 2007-08-30 Dsm Fine Chemicals Austria Nfg Gmbh & Co Kg Process for preparing 3, 4-disubstituted phenylacetic acids and novel intermediates
WO2007128761A2 (de) 2006-05-04 2007-11-15 Boehringer Ingelheim International Gmbh Verwendungen von dpp iv inhibitoren
US7390908B2 (en) 2001-08-17 2008-06-24 Astrazeneca Ab Compounds effecting glucokinase
WO2008078674A1 (ja) 2006-12-25 2008-07-03 Kyorin Pharmaceutical Co., Ltd. グルコキナーゼ活性化物質
US7541373B2 (en) 2002-06-27 2009-06-02 Novo Nordisk A/S Aryl carbonyl derivatives as therapeutic agents
WO2009091014A1 (ja) 2008-01-18 2009-07-23 Astellas Pharma Inc. フェニルアセトアミド誘導体
WO2009133687A1 (ja) 2008-04-28 2009-11-05 杏林製薬株式会社 シクロペンチルアクリル酸アミド誘導体
WO2010029461A1 (en) * 2008-09-11 2010-03-18 Pfizer Inc. Heteroaryls amide derivatives and their use as glucokinase activators
WO2010021882A3 (en) * 2008-08-19 2010-05-14 Janssen Pharmaceutica Nv Cold menthol receptor antagonists
US7741327B2 (en) 2008-04-16 2010-06-22 Hoffmann-La Roche Inc. Pyrrolidinone glucokinase activators
US7745491B2 (en) 2004-08-12 2010-06-29 Prosidion Limited Substituted phenylacetamides and their use as glucokinase activators
US7888504B2 (en) 2006-07-06 2011-02-15 Bristol-Myers Squibb Company Glucokinase activators and methods of using same
US7910747B2 (en) 2006-07-06 2011-03-22 Bristol-Myers Squibb Company Phosphonate and phosphinate pyrazolylamide glucokinase activators
US7935699B2 (en) 2006-07-24 2011-05-03 Hoffmann-La Roche Inc. Pyrazole glucokinase activators
WO2011073117A1 (en) * 2009-12-14 2011-06-23 F. Hoffmann-La Roche Ag Azaindole glucokinase activators
US8008332B2 (en) 2006-05-31 2011-08-30 Takeda San Diego, Inc. Substituted indazoles as glucokinase activators
WO2011123572A1 (en) 2010-03-31 2011-10-06 The Scripps Research Institute Reprogramming cells
US8034822B2 (en) 2006-03-08 2011-10-11 Takeda San Diego, Inc. Glucokinase activators
US8124617B2 (en) 2005-09-01 2012-02-28 Takeda San Diego, Inc. Imidazopyridine compounds
US8163779B2 (en) 2006-12-20 2012-04-24 Takeda San Diego, Inc. Glucokinase activators
US8173645B2 (en) 2007-03-21 2012-05-08 Takeda San Diego, Inc. Glucokinase activators
US8258134B2 (en) 2008-04-16 2012-09-04 Hoffmann-La Roche Inc. Pyridazinone glucokinase activators
AU2005301608B2 (en) * 2004-11-02 2012-09-13 Msd K.K. Aryloxy-substituted benzimidazole derivatives
US8455496B2 (en) 2009-03-11 2013-06-04 Pfizer Inc. Benzofuranyl derivatives
WO2014114825A1 (es) 2013-01-22 2014-07-31 Consejo Superior De Investigaciones Científicas (Csic) Benzotiazoles sustituidos y sus aplicaciones terapeuticas para el tratamiento de enfermedades humanas
USRE45183E1 (en) 2004-01-06 2014-10-07 Novo Nordisk A/S Heteroaryl-ureas and their use as glucokinase activators
EP2878339A1 (en) 2013-12-02 2015-06-03 Siena Biotech S.p.A. SIP3 antagonists
US11229636B2 (en) 2015-05-15 2022-01-25 Aurigene Discovery Technologies Limited Substituted tetrahydroquinolinone compounds as ROR gamma modulators
US11833136B2 (en) 2018-06-12 2023-12-05 Vtv Therapeutics Llc Therapeutic uses of glucokinase activators in combination with insulin or insulin analogs

Families Citing this family (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6775812B2 (en) * 2002-07-17 2004-08-10 Hewlett-Packard Development Company, L.P. Layout design process and system for providing bypass capacitance and compliant density in an integrated circuit
WO2004031179A1 (en) 2002-10-03 2004-04-15 F. Hoffmann-La Roche Ag Indole-3-carboxamides as glucokinase (gk) activators
GB0226930D0 (en) 2002-11-19 2002-12-24 Astrazeneca Ab Chemical compounds
MY141521A (en) 2002-12-12 2010-05-14 Hoffmann La Roche 5-substituted-six-membered heteroaromatic glucokinase activators
WO2004072066A1 (en) * 2003-02-11 2004-08-26 Prosidion Limited Tri(cyclo) substituted amide glucokinase activator compounds
DE602004012858T2 (de) * 2003-06-20 2009-05-14 F. Hoffmann-La Roche Ag 2-aminobenzothiazole als cb1 rezeptor inverse agonisten
AU2005214137B2 (en) * 2004-02-18 2008-05-29 Astrazeneca Ab Compounds
KR20070007103A (ko) * 2004-02-18 2007-01-12 아스트라제네카 아베 벤즈아미드 유도체 및 이의 글루코키나아제 활성화제로서의용도
TW200600086A (en) * 2004-06-05 2006-01-01 Astrazeneca Ab Chemical compound
WO2006040527A1 (en) 2004-10-16 2006-04-20 Astrazeneca Ab Process for making phenoxy benzamide compounds
GB0423043D0 (en) * 2004-10-16 2004-11-17 Astrazeneca Ab Compounds
GB0423044D0 (en) * 2004-10-16 2004-11-17 Astrazeneca Ab Compounds
DE102005012872A1 (de) * 2005-03-19 2006-09-28 Sanofi-Aventis Deutschland Gmbh Substituierte, bizyklische 8-Pyrrolidino-benzimidazole, Verfahren zu ihrer Herstellung und ihre Verwendung als Arzneimittel
JP2008542247A (ja) * 2005-05-24 2008-11-27 アストラゼネカ アクチボラグ グルコキナーゼモジュレーターとしての2−フェニル置換イミダゾール[4,5b]ピリジン/ピラジンおよびプリン誘導体
TW200714597A (en) * 2005-05-27 2007-04-16 Astrazeneca Ab Chemical compounds
US20080234273A1 (en) * 2005-07-09 2008-09-25 Mckerrecher Darren Heteroaryl Benzamide Derivatives for Use as Glk Activators in the Treatment of Diabetes
NZ575514A (en) * 2005-07-09 2009-11-27 Astrazeneca Ab Heteroaryl benzamide derivatives for use as GLK activators in the treatment of diabetes
JP2009500442A (ja) * 2005-07-09 2009-01-08 アストラゼネカ アクチボラグ 2型糖尿病を処置するためのグルコキナーゼのモジュレーターとしての2−ヘテロシクリルオキシベンゾイルアミノヘテロシクリル化合物
AU2006267338B2 (en) 2005-07-13 2012-08-16 Msd K.K. Heterocycle-substituted benzimidazole derivative
US9202182B2 (en) * 2005-08-11 2015-12-01 International Business Machines Corporation Method and system for analyzing business architecture
GT200600428A (es) * 2005-09-30 2007-05-21 Compuestos organicos
GT200600429A (es) * 2005-09-30 2007-04-30 Compuestos organicos
US20090005391A1 (en) * 2005-11-03 2009-01-01 Matthew Colin Thor Fyfe Tricyclo Substituted Amides
US20070197532A1 (en) * 2005-11-18 2007-08-23 Cao Sheldon X Glucokinase activators
TW200738621A (en) * 2005-11-28 2007-10-16 Astrazeneca Ab Chemical process
TW200825063A (en) * 2006-10-23 2008-06-16 Astrazeneca Ab Chemical compounds
SA07280576B1 (ar) * 2006-10-26 2011-06-22 استرازينيكا ايه بي مركبات بنزويل أمينو سيكليل غير متجانسة بأعتبارها عوامل منشطة للجلوكوكيناز
BRPI0721143A2 (pt) * 2006-12-21 2014-03-11 Astrazeneca Ab Forma cristalina do composto processo para a formação da mesma, uso de um composto, método para tratar doenças mediadas por ativador de glicocinase
BRPI0808267A2 (pt) 2007-03-07 2014-07-22 Kyorin Phamaceutical Co., Ltd "composto representado pela formula geral (1) ou um sal farmeceuticamente aceitável do mesmo; método para o tratamento ou prevenção de diabetes ; uso do composto; composição farmacêutica e composto representado pela formula geral (3)".
CL2009000004A1 (es) * 2008-01-15 2010-02-19 Lilly Co Eli Forma cristalina de r-2-(4-ciclopropansulfonil-fenil)-n-pirazin-2-il-3-(tetrahidropiran-4-il)-propionamida; composicion farmaceutica que comprende a dicha forma cristalina; y uso para el tratamiento de diabetes o hiperglicemia.
EP2324028A2 (en) 2008-08-04 2011-05-25 AstraZeneca AB Therapeutic agents 414
GB0902406D0 (en) * 2009-02-13 2009-04-01 Astrazeneca Ab Crystalline polymorphic form
GB0902434D0 (en) * 2009-02-13 2009-04-01 Astrazeneca Ab Chemical process
AR076220A1 (es) 2009-04-09 2011-05-26 Astrazeneca Ab Derivados de pirazol [4,5 - e] pirimidina
AR076221A1 (es) 2009-04-09 2011-05-26 Astrazeneca Ab Derivado de pirazol [4,5-e] pirimidina y su uso para tratar diabetes y obesidad
US20130156720A1 (en) 2010-08-27 2013-06-20 Ironwood Pharmaceuticals, Inc. Compositions and methods for treating or preventing metabolic syndrome and related diseases and disorders
US9616097B2 (en) 2010-09-15 2017-04-11 Synergy Pharmaceuticals, Inc. Formulations of guanylate cyclase C agonists and methods of use
CN108676076A (zh) 2011-03-01 2018-10-19 辛纳吉制药公司 制备鸟苷酸环化酶c激动剂的方法
JP6606491B2 (ja) 2013-06-05 2019-11-13 シナジー ファーマシューティカルズ インコーポレイテッド グアニル酸シクラーゼcの超高純度アゴニスト、その作成および使用方法
JP6769963B2 (ja) 2014-08-29 2020-10-14 ティエエッセ ファルマ ソチエタ レスポンサビリタ リミタータ α−アミノ−β−カルボキシムコン酸セミアルデヒド脱炭酸酵素の阻害剤
AU2017342083A1 (en) 2016-10-14 2019-04-11 Tes Pharma S.R.L. Inhibitors of alpha-amino-beta-carboxymuconic acid semialdehyde decarboxylase
KR20210111248A (ko) 2018-11-20 2021-09-10 테스 파마 에스.알.엘. α-아미노-β-카르복시뮤콘산 세미알데하이드 데카르복실라제의 저해제

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000058293A2 (en) * 1999-03-29 2000-10-05 F. Hoffmann-La Roche Ag Glucokinase activators

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3840550A (en) * 1971-05-24 1974-10-08 Ciba Geigy Corp Certain 6-isothiocyanobenzothiazoles
US6320050B1 (en) * 1999-03-29 2001-11-20 Hoffmann-La Roche Inc. Heteroaromatic glucokinase activators

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000058293A2 (en) * 1999-03-29 2000-10-05 F. Hoffmann-La Roche Ag Glucokinase activators

Cited By (90)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7199140B2 (en) 2001-06-26 2007-04-03 Astrazeneca Ab Vinyl phenyl derivatives as GLK activators
US7390908B2 (en) 2001-08-17 2008-06-24 Astrazeneca Ab Compounds effecting glucokinase
USRE45670E1 (en) 2002-06-27 2015-09-15 Novo Nordisk A/S Aryl carbonyl derivatives as therapeutic agents
WO2004002481A1 (en) * 2002-06-27 2004-01-08 Novo Nordisk A/S Aryl carbonyl derivatives as therapeutic agents
US7897628B2 (en) 2002-06-27 2011-03-01 Novo Nordisk A/S Aryl carbonyl derivatives as therapeutic agents
US8063081B2 (en) 2002-06-27 2011-11-22 Novo Nordisk A/S Aryl carbonyl derivatives as therapeutic agents
JP4881559B2 (ja) * 2002-06-27 2012-02-22 ノボ・ノルデイスク・エー/エス 治療薬としてのアリールカルボニル誘導体
EP2471533A1 (en) * 2002-06-27 2012-07-04 Novo Nordisk A/S Aryl carbonyl derivatives as therapeutic agents
US7541373B2 (en) 2002-06-27 2009-06-02 Novo Nordisk A/S Aryl carbonyl derivatives as therapeutic agents
JP2005537333A (ja) * 2002-06-27 2005-12-08 ノボ・ノルデイスク・エー/エス 治療薬としてのアリールカルボニル誘導体
AU2003243921B2 (en) * 2002-06-27 2009-05-07 Novo Nordisk A/S Aryl carbonyl derivatives as therapeutic agents
US7384967B2 (en) 2002-06-27 2008-06-10 Novo Nordisk A/S Aryl carbonyl derivatives as therapeutic agents
AU2003302279B2 (en) * 2002-10-03 2008-06-12 Novartis Ag Substituted (thiazol-2-yl) -amide or sulfonamide as glycokinase activators useful in the treatment of Type 2 diabetes
JP2006509774A (ja) * 2002-10-03 2006-03-23 ノバルティス アクチエンゲゼルシャフト 2型糖尿病の処置において有用なグルコキナーゼアクチベーターとしての置換(チアゾール−2−イル)−アミドまたはスルホンアミド
US7812167B2 (en) 2002-10-03 2010-10-12 Novartis, Ag Substituted (thiazol-2-yl)-amides or sulfonamides as glucokinase activators useful in the treatment of type 2 diabetes
WO2004050645A1 (en) * 2002-10-03 2004-06-17 Novartis Ag Substituted (thiazol-2-yl) -amide or sulfonamide as glycokinase activators useful in the treatment of type 2 diabetes
EP1549626A1 (en) * 2002-10-03 2005-07-06 Novartis AG Substituted (thiazol-2-yl) -amide or sulfonamide as glycokinase activators useful in the treatment of type 2 diabetes
WO2004045614A1 (en) * 2002-11-19 2004-06-03 Astrazeneca Ab Quinoline derivatives as glucokinase ligands
US7230108B2 (en) 2002-11-19 2007-06-12 Astrazeneca Ab Quinoline derivatives as glucokinase ligands
US7214681B2 (en) 2003-02-11 2007-05-08 Prosidion Limited Tri(cyclo) substituted amide compounds
JP2006517590A (ja) * 2003-02-11 2006-07-27 プロシディオン・リミテッド トリ(シクロ)置換されたアミド化合物
JP4757188B2 (ja) * 2003-02-11 2011-08-24 プロシディオン・リミテッド トリ(シクロ)置換されたアミド化合物
US7582632B2 (en) 2003-02-11 2009-09-01 Prosidion Limited Tri(cyclo) substituted amide compounds
USRE45183E1 (en) 2004-01-06 2014-10-07 Novo Nordisk A/S Heteroaryl-ureas and their use as glucokinase activators
JP4700684B2 (ja) * 2004-04-02 2011-06-15 ノバルティス アーゲー 2型糖尿病の処置に有用なグルコキナーゼアクティベーターとしてのスルホンアミド−チアゾロピリジン誘導体
AU2005229415B2 (en) * 2004-04-02 2009-05-14 Novartis Ag Thiazolopyridine derivatives, pharmaceutical conditions containing them and methods of treating glucokinase mediated conditions
KR100830007B1 (ko) * 2004-04-02 2008-05-15 노파르티스 아게 2형 당뇨병 치료에 유용한 글루코키나제 활성화제로서의술폰아미드-티아졸피리딘 유도체
JP2007530631A (ja) * 2004-04-02 2007-11-01 ノバルティス アクチエンゲゼルシャフト チアゾロピリジン誘導体、それを含む医薬組成物およびグルコキナーゼ介在性状態の処置法
JP2007530632A (ja) * 2004-04-02 2007-11-01 ノバルティス アクチエンゲゼルシャフト 2型糖尿病の処置に有用なグルコキナーゼアクティベーターとしてのスルホンアミド−チアゾロピリジン誘導体
WO2005095417A1 (en) * 2004-04-02 2005-10-13 Novartis Ag Thiazolopyridine derivates, pharmaceutical conditions containing them and methods of treating glucokinase mediated conditions
US7750020B2 (en) 2004-04-02 2010-07-06 Novartis Ag Sulfonamide-thiazolpyridine derivatives as glucokinase activators useful the treatment of Type 2 diabetes
WO2005095418A1 (en) * 2004-04-02 2005-10-13 Novartis Ag Sulfonamide-thiazolpyridine derivatives as glucokinase activators useful the treatment of type 2 diabetes
WO2005103021A1 (en) * 2004-04-21 2005-11-03 Prosidion Limited Tri(cyclo) substituted amide compounds
EA012204B1 (ru) * 2004-04-21 2009-08-28 Прозидион Лимитед Три(цикло)замещённые амидные соединения
US7745491B2 (en) 2004-08-12 2010-06-29 Prosidion Limited Substituted phenylacetamides and their use as glucokinase activators
AU2005301608B2 (en) * 2004-11-02 2012-09-13 Msd K.K. Aryloxy-substituted benzimidazole derivatives
US8124617B2 (en) 2005-09-01 2012-02-28 Takeda San Diego, Inc. Imidazopyridine compounds
WO2007039177A2 (en) 2005-09-29 2007-04-12 Sanofi-Aventis Phenyl- and pyridinyl- 1, 2 , 4 - oxadiazolone derivatives, processes for their preparation and their use as pharmaceuticals
WO2007096034A1 (en) * 2006-02-22 2007-08-30 Dsm Fine Chemicals Austria Nfg Gmbh & Co Kg Process for preparing 3, 4-disubstituted phenylacetic acids and novel intermediates
US8034822B2 (en) 2006-03-08 2011-10-11 Takeda San Diego, Inc. Glucokinase activators
EP2351568A2 (de) 2006-05-04 2011-08-03 Boehringer Ingelheim International GmbH Verwendungen von dpp iv Inhibitoren
WO2007128761A2 (de) 2006-05-04 2007-11-15 Boehringer Ingelheim International Gmbh Verwendungen von dpp iv inhibitoren
US8008332B2 (en) 2006-05-31 2011-08-30 Takeda San Diego, Inc. Substituted indazoles as glucokinase activators
US8394843B2 (en) 2006-05-31 2013-03-12 Takeda California, Inc. Substituted isoindoles as glucokinase activators
US8153677B2 (en) 2006-07-06 2012-04-10 Bristol-Myers Squibb Company Substituted pyrazolylamide compounds useful as glucokinase activators
US7910747B2 (en) 2006-07-06 2011-03-22 Bristol-Myers Squibb Company Phosphonate and phosphinate pyrazolylamide glucokinase activators
US8614332B2 (en) 2006-07-06 2013-12-24 Bristol-Myers Squibb Company Substituted pyrazolylamides useful as glucokinase activators
US7888504B2 (en) 2006-07-06 2011-02-15 Bristol-Myers Squibb Company Glucokinase activators and methods of using same
US7935699B2 (en) 2006-07-24 2011-05-03 Hoffmann-La Roche Inc. Pyrazole glucokinase activators
US8163779B2 (en) 2006-12-20 2012-04-24 Takeda San Diego, Inc. Glucokinase activators
US8173649B2 (en) 2006-12-25 2012-05-08 Kyorin Pharmaceutical Co., Ltd. Glucokinase activator
WO2008078674A1 (ja) 2006-12-25 2008-07-03 Kyorin Pharmaceutical Co., Ltd. グルコキナーゼ活性化物質
US8173645B2 (en) 2007-03-21 2012-05-08 Takeda San Diego, Inc. Glucokinase activators
US8329707B2 (en) 2008-01-18 2012-12-11 Astellas Pharma Inc. Substituted pyrazine compounds
WO2009091014A1 (ja) 2008-01-18 2009-07-23 Astellas Pharma Inc. フェニルアセトアミド誘導体
US8258134B2 (en) 2008-04-16 2012-09-04 Hoffmann-La Roche Inc. Pyridazinone glucokinase activators
US7741327B2 (en) 2008-04-16 2010-06-22 Hoffmann-La Roche Inc. Pyrrolidinone glucokinase activators
US9452977B2 (en) 2008-04-28 2016-09-27 Kyorin Pharmaceutical Co., Ltd. Cyclopentylacrylamide derivative
WO2009133687A1 (ja) 2008-04-28 2009-11-05 杏林製薬株式会社 シクロペンチルアクリル酸アミド誘導体
US8946440B2 (en) 2008-04-28 2015-02-03 Kyorin Pharmaceutical Co., Ltd. Cyclopentylacrylamide derivative
US8653099B2 (en) 2008-08-19 2014-02-18 Janssen Pharmaceutica Cold menthol receptor antagonists
US9096521B2 (en) 2008-08-19 2015-08-04 Janssen Pharmaceutica Nv Cold menthol receptor antagonists
WO2010021882A3 (en) * 2008-08-19 2010-05-14 Janssen Pharmaceutica Nv Cold menthol receptor antagonists
US8389552B2 (en) 2008-09-11 2013-03-05 Pfizer Inc. (S)-6-(2-(4-(cyclobutylsulfonyl)-1H-imidazol-1-yl)-3-cyclopentylpropanamido)nicotinic acid useful as a glucokinase activator
KR101258331B1 (ko) 2008-09-11 2013-04-26 화이자 인코포레이티드 헤테로아릴 아미드 유도체 및 글루코키나제 활성화제로서의 그의 용도
WO2010029461A1 (en) * 2008-09-11 2010-03-18 Pfizer Inc. Heteroaryls amide derivatives and their use as glucokinase activators
US7977367B2 (en) 2008-09-11 2011-07-12 Pfizer Inc Substituted imidazole propanamide glucokinase activators
TWI382026B (zh) * 2008-09-11 2013-01-11 Pfizer 經取代的雜芳基物
US8329920B2 (en) 2008-09-11 2012-12-11 Pfizer Inc. Substituted imidazoles useful for treating type II diabetes
US8455496B2 (en) 2009-03-11 2013-06-04 Pfizer Inc. Benzofuranyl derivatives
US8735396B2 (en) 2009-03-11 2014-05-27 Pfizer Inc. Benzofuranyl derivatives
CN102686587A (zh) * 2009-12-14 2012-09-19 霍夫曼-拉罗奇有限公司 氮杂吲哚葡糖激酶激活剂
WO2011073117A1 (en) * 2009-12-14 2011-06-23 F. Hoffmann-La Roche Ag Azaindole glucokinase activators
CN102686587B (zh) * 2009-12-14 2015-05-27 霍夫曼-拉罗奇有限公司 氮杂吲哚葡糖激酶激活剂
US8222416B2 (en) 2009-12-14 2012-07-17 Hoffmann-La Roche Inc. Azaindole glucokinase activators
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
EP3348550A1 (en) 2013-01-22 2018-07-18 Consejo Superior de Investigaciones Cientificas (CSIC) Substituted benzothiazoles and therapeutic uses thereof for the treatment of human diseases
US9592224B2 (en) 2013-01-22 2017-03-14 Donsejo Superior De Investigaciones Científicas (Csic) Substituted benzothiazoles and therapeutic uses thereof for the treatment of human diseases
WO2014114825A1 (es) 2013-01-22 2014-07-31 Consejo Superior De Investigaciones Científicas (Csic) Benzotiazoles sustituidos y sus aplicaciones terapeuticas para el tratamiento de enfermedades humanas
WO2015082357A1 (en) 2013-12-02 2015-06-11 Siena Biotech S.P.A. S1p3 antagonists
US9951017B2 (en) 2013-12-02 2018-04-24 Teva Pharmaceutical Industries Limited S1P3 antagonists
EP2878339A1 (en) 2013-12-02 2015-06-03 Siena Biotech S.p.A. SIP3 antagonists
EP3689864A1 (en) 2013-12-02 2020-08-05 Teva Pharmaceutical Industries Limited S1p3 antagonists
EP3896068A1 (en) 2013-12-02 2021-10-20 Teva Pharmaceutical Industries Limited S1p3 antagonists
US11472772B2 (en) 2013-12-02 2022-10-18 Teva Pharmaceutical Industries Limited S1P3 antagonists
US11229636B2 (en) 2015-05-15 2022-01-25 Aurigene Discovery Technologies Limited Substituted tetrahydroquinolinone compounds as ROR gamma modulators
US11833136B2 (en) 2018-06-12 2023-12-05 Vtv Therapeutics Llc Therapeutic uses of glucokinase activators in combination with insulin or insulin analogs
US11974989B2 (en) 2018-06-12 2024-05-07 Vtv Therapeutics Llc Therapeutic uses of glucokinase activators in combination with insulin or insulin analogs

Also Published As

Publication number Publication date
US20020103241A1 (en) 2002-08-01
DK1341774T3 (da) 2006-06-12
DE60117059D1 (de) 2006-04-13
BR0115999A (pt) 2003-09-30
ATE316965T1 (de) 2006-02-15
US20020103199A1 (en) 2002-08-01
CA2429642A1 (en) 2002-06-13
ZA200303748B (en) 2004-08-16
ES2256340T3 (es) 2006-07-16
DE60117059T2 (de) 2006-10-26
AR031627A1 (es) 2003-09-24
JP4109111B2 (ja) 2008-07-02
MXPA03005119A (es) 2004-10-15
CN1476438A (zh) 2004-02-18
AU2190202A (en) 2002-06-18
US6448399B1 (en) 2002-09-10
CN1226294C (zh) 2005-11-09
JP2004517087A (ja) 2004-06-10
EP1341774B1 (en) 2006-02-01
CA2429642C (en) 2007-11-20
KR20030068555A (ko) 2003-08-21
US6441184B1 (en) 2002-08-27
US6545155B2 (en) 2003-04-08
PE20020753A1 (es) 2002-08-27
PT1341774E (pt) 2006-05-31
KR100545431B1 (ko) 2006-01-24
US20020107396A1 (en) 2002-08-08
EP1341774A1 (en) 2003-09-10
GT200100243A (es) 2002-06-04
UY27055A1 (es) 2002-07-31
AU2002221902B2 (en) 2006-11-23
PA8534301A1 (es) 2002-08-26

Similar Documents

Publication Publication Date Title
EP1341774B1 (en) Fused heteroaromatic glucokinase activators
AU2002221902A1 (en) Fused heteroaromatic glucokinase activators
US6433188B1 (en) Fused heteroaromatic glucokinase activators
EP1349856B1 (en) Isoindolin-1-one glucokinase activators
EP1311504B1 (en) Tetrazolyl-phenyl acetamide glucokinase activators
EP1282611B1 (en) Substituted phenylacetamides and their use as glucokinase activators
EP1283830B1 (en) Para-amine substituted phenylamide glucokinase activators
KR100502032B1 (ko) 트랜스 올레핀 글루코키나제 활성화제
AU2002238415A1 (en) Isoindolin-1-one glucokinase activators
AU2001283998A1 (en) Tetrazolyl-phenyl acetamide glucokinase activators
AU2001265914A1 (en) Para-amine substituted phenylamide glucokinase activators

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PH PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2003/03748

Country of ref document: ZA

Ref document number: 200303748

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 2429642

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 018194494

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2002221902

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2001999565

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1020037007517

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2002547912

Country of ref document: JP

WWP Wipo information: published in national office

Ref document number: 1020037007517

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2001999565

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWG Wipo information: grant in national office

Ref document number: 1020037007517

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 2001999565

Country of ref document: EP