WO2007041366A1 - Sulfonamide derivatives as glycokinase activators useful in the treatment of type 2 diabetes - Google Patents

Sulfonamide derivatives as glycokinase activators useful in the treatment of type 2 diabetes Download PDF

Info

Publication number
WO2007041366A1
WO2007041366A1 PCT/US2006/038201 US2006038201W WO2007041366A1 WO 2007041366 A1 WO2007041366 A1 WO 2007041366A1 US 2006038201 W US2006038201 W US 2006038201W WO 2007041366 A1 WO2007041366 A1 WO 2007041366A1
Authority
WO
WIPO (PCT)
Prior art keywords
ethyl
methoxy
cyclopentyl
ylcarbamoyl
amino
Prior art date
Application number
PCT/US2006/038201
Other languages
French (fr)
Inventor
Gregory Raymond Bebernitz
Louise Kirman
Original Assignee
Novartis Ag
Novartis Pharma Gmbh
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 Novartis Ag, Novartis Pharma Gmbh filed Critical Novartis Ag
Priority to CA002623045A priority Critical patent/CA2623045A1/en
Priority to US12/088,594 priority patent/US8252931B2/en
Priority to EP06815877A priority patent/EP1934233B1/en
Priority to BRPI0616710-1A priority patent/BRPI0616710A2/en
Priority to JP2008533703A priority patent/JP2009510111A/en
Priority to AU2006297128A priority patent/AU2006297128B2/en
Priority to ES06815877T priority patent/ES2385052T3/en
Publication of WO2007041366A1 publication Critical patent/WO2007041366A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole 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
    • C07D231/38Nitrogen atoms
    • C07D231/40Acylated on said nitrogen atom
    • 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
    • A61K31/4151,2-Diazoles
    • 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
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen 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
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more 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, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/78Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 2
    • C07D239/84Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/10Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D241/14Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having 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
    • C07D241/20Nitrogen atoms
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D285/00Heterocyclic compounds containing rings having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by groups C07D275/00 - C07D283/00
    • C07D285/01Five-membered rings
    • C07D285/02Thiadiazoles; Hydrogenated thiadiazoles
    • C07D285/04Thiadiazoles; Hydrogenated thiadiazoles not condensed with other rings
    • C07D285/121,3,4-Thiadiazoles; Hydrogenated 1,3,4-thiadiazoles
    • C07D285/1251,3,4-Thiadiazoles; Hydrogenated 1,3,4-thiadiazoles with oxygen, sulfur or nitrogen atoms, directly attached to ring carbon atoms, the nitrogen atoms not forming part of a nitro radical
    • C07D285/135Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D285/00Heterocyclic compounds containing rings having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by groups C07D275/00 - C07D283/00
    • C07D285/15Six-membered rings
    • C07D285/16Thiadiazines; Hydrogenated thiadiazines
    • 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
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems

Definitions

  • the present invention relates to certain sulfonamide derivatives, pharmaceutical compositions containing them, and to methods of treating glucokinase mediated conditions, in particular, impaired glucose tolerance and type 2 diabetes, by employing such compounds.
  • R 1 and R 2 are, independently from each other, hydrogen, halogen, cyano, nitro, optionally substituted alkyl, alkoxy, alkylthio, alkylthiono, sulfonyl, free or esterified carboxy, carbamoyl, sulfamoyl, optionally substituted amino, aryl or heterocyclyl; or
  • R 2 is absent
  • R 3 is (C 3 . 6 )cycloalkyl or (C 3-6 )heterocyclyl;
  • R 4 is hydrogen, halogen, cyano, lower alkyl or lower alkoxy
  • R 5 is (C 3-12 )cycloalkyl, (C 6 .io)aryl, (C 3-10 )heterocyclyl or (d. 6 )alkyl optionally substituted by (C 1-6 )alkoxy, (C 1-2 )alkoxy-(Ci -4 )alkoxy, (C 1-6 )alkylthio, (C 3-7 )cycloalkyl, (C 3-7 )cycloalkoxy, (C 3-7 )cycloalkylthio, (C 6-10 )aryl, (C 6- i 0 )aryloxy, (C 6-10 )arylthio, (C 3-1 o)heterocyclyl or (C 3-10 )heterocyclyloxy;
  • R 6 is free or esterified carboxy, tetrazolyl, cyano or -C(O)NR 7 R 8 in which
  • R 7 and R 8 are, independently from each other, hydrogen, optionally substituted alkyl, cycloalkyl, aryl or heterocyclyl; or R 7 and R 8 combined are alkylene which together with the nitrogen atom to which they are attached form a 4- to 7-membered ring; n is an integer from 1 to 6; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
  • the compounds of the present invention provide pharmacological agents which are glucokinase activators and, thus, may be employed for the treatment of glucokinase mediated conditions. Accordingly, the compounds of formula (I) may be employed for prevention and treatment of impaired glucose tolerance, type 2 diabetes and obesity.
  • optionally substituted alkyl refers to unsubstituted or substituted alkyl groups, i.e., straight- or branched-chain hydrocarbon groups having 1-20 carbon atoms, preferably 1-10 carbon atoms.
  • exemplary unsubstituted alkyl groups include methyl, ethyl, propyl, isopropyl, /7-butyl, f-butyl, isobutyl, pentyl, hexyl, isohexyl, heptyl, 4,4-dimethylpentyl, octyl and the like.
  • Substituted alkyl groups include, but are not limited to, alkyl groups substituted by one or more of the following groups: halogen, hydroxy, alkanoyl, alkoxy, alkanoyloxy, thiol, alkylthio, alkylthiono, sulfonyl, sulfamoyl, carbamoyl, cyano, carboxy, acyl, aryl, alkenyl, alkynyl, aralkoxy, guanidino, optionally substituted amino, heterocyclyl including imidazolyl, furyl, thienyl, thiazolyl, pyrrolidyl, pyridyl, pyrimidyl and the like.
  • lower alkyl refers to those alkyl groups as described above having 1-7, preferably 2-4 carbon atoms.
  • halogen refers to fluorine, chlorine, bromine and iodine.
  • alkenyl refers to any of the above alkyl groups having at least two carbon atoms and further containing a carbon to carbon double bond at the point of attachment. Groups having 2-4 carbon atoms are preferred.
  • alkynyl refers to any of the above alkyl groups having at least two carbon atoms and further containing a carbon to carbon triple bond at the point of attachment. Groups having 2-4 carbon atoms are preferred.
  • alkylene refers to a straight-chain bridge of 3-6 carbon atoms connected by single bonds, e.g., -(CH 2 ) X -, wherein x is 3-6, which may be interrupted with one or more heteroatoms selected from O, -O-C(O)-, S, S(O), S(O) 2 or NR, wherein R may be hydrogen, alkyl, cycloalkyl, aryl, heterocyclyl, aralkyl, heteroaralkyl, acyl, carbamoyl, sulfonyl, alkoxycarbonyl, aryloxycarbonyl or aralkoxycarbonyl and the like; and the alkylene may further be substituted with one or more substituents selected from optionally substituted alkyl, cycloalkyl, aryl, heterocyclyl, oxo, halogen, hydroxy, carboxy, alkoxy, alkoxycarbonyl and the like.
  • cycloalkyl refers to optionally substituted monocyclic, bicyclic or tricyclic hydrocarbon groups of 3-12 carbon atoms, each of which may contain one or more carbon to carbon double bonds, or the cycloalkyl may be substituted by one or more substituents, such as alkyl, halo, oxo, hydroxy, alkoxy, alkanoyl, acylamino, carbamoyl, alkylamino, dialkylamino, thiol, alkylthio, cyano, carboxy, alkoxycarbonyl, sulfonyl, sulfonamido, sulfamoyl, heterocyclyl and the like.
  • substituents such as alkyl, halo, oxo, hydroxy, alkoxy, alkanoyl, acylamino, carbamoyl, alkylamino, dialkylamino, thiol, alkylthio, cyano, carboxy
  • Exemplary monocyclic hydrocarbon groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl and cyclohexenyl and the like.
  • bicyclic hydrocarbon groups include bornyl, indyl, hexahydroindyl, tetrahydronaphthyl, decahydronaphthyl, bicyclo[2.1.1]hexyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.1 ]heptenyl, 6,6-dimethylbicyclo[3.1.1 ]heptyl, 2,6,6-trimethylbicyclo[3.1.1 ]heptyl, bicyclo[2.2.2]octyl and the like.
  • Exemplary tricyclic hydrocarbon groups include adamantyl and the like.
  • alkoxy refers to alkyl-O-.
  • alkanoyl refers to alkyl-C(O)-.
  • alkanoyloxy refers to alkyl-C(O)-O-.
  • alkylamino and “dialkylamino” refer to alkyl-NH- and (alkyl) 2 N-, respectively. - A -
  • alkanoylamino refers to alkyl-C(O)-NH-.
  • alkylthio refers to alkyl-S-.
  • alkylthiono refers to alkyl-S(O)-.
  • alkylsulfonyl refers to alkyl-S(O) 2 -.
  • alkoxycarbonyl refers to alkyl-O-C(O)-.
  • alkoxycarbonyloxy refers to alkyl-O-C(O)O-.
  • carbamoyl refers to H 2 NC(O)-, alkyl-NHC(O)-, (alkyl) 2 NC(O)-, aryl-NHC(O)-, alkyl(aryl)-NC(O)-, heteroaryl-N HC(O)-, alkyl(heteroaryl)-NC(O)-, aralkyl-NHC(O)-, alkyl(aralkyl)-NC(O)- and the like.
  • sulfamoyl refers to H 2 NS(O) 2 -, alkyl-NHS(O) 2 -, (alkyl) 2 NS(O) 2 -, aryl-NHS(O) 2 -, alkyl(aryl)-NS(O) 2 -, (aryl) 2 NS(O) 2 -, heteroaryl-NHS(O) 2 -, aralkyl-NHS(O) 2 -, heteroaralkyl- NHS(O) 2 - and the like.
  • sulfonamido refers to alkyl-S(O) 2 -NH-, ary 1-S(O) 2 -N H-, aralkyl-S(O) 2 -NH-, heteroaryl-S(O) 2 -NH-, heteroaralkyl-S(O) 2 -NH-, alkyl-S(O) 2 -N(alkyl)-, aryl-S(O) 2 -N(alkyl)-, aralkyl-S(O) 2 -N(alkyl)-, heteroaryl-S(O) 2 -N(alkyl)-, heteroaralkyl-S(O) 2 -N(alkyl)- and the like.
  • sulfonyl refers to alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aralkylsulfonyl, heteroaralkylsulfonyl and the like.
  • optionally substituted amino refers to an amino group which may optionally be substituted by substituents such as optionally substituted alkyl, acyl, sulfonyl, alkoxycarbonyl, cycloalkoxycarbonyl, aryloxycarbonyl, heteroaryloxycarbonyl, aralkoxycarbonyl, heteroaralkoxycarbonyl, carbamoyl and the like.
  • aryl refers to monocyclic or bicyclic aromatic hydrocarbon groups having 6-12 carbon atoms in the ring portion, such as phenyl, biphenyl, naphthyl and tetrahydronaphthyl, each of which may optionally be substituted by 1-4 substituents, such as optionally substituted alkyl, trifluoromethyl, cycloalkyl, halo, hydroxy, alkoxy, acyl, alkanoyloxy, aryloxy, optionally substituted amino, thiol, alkylthio, arylthio, nitro, cyano, carboxy, alkoxycarbonyl, carbamoyl, alkylthiono, sulfonyl, sulfonamido, heterocyclyl and the like.
  • monocyclic aryl refers to optionally substituted phenyl as described under aryl.
  • aralkyl refers to an aryl group bonded directly through an alkyl group, such as benzyl.
  • aralkanoyl refers to aralkyl-C(O)-.
  • aralkylthio refers to aralkyl-S-.
  • alkoxy refers to an aryl group bonded directly through an alkoxy group.
  • arylsulfonyl refers to aryl-S(O) 2 -.
  • arylthio refers to aryl-S-.
  • aroyl refers to aryl-C(O)-.
  • aroyloxy refers to aryl-C(O)-O-.
  • aroylamino refers to aryl-C(O)-NH-.
  • aryloxycarbonyl refers to aryl-O-C(O)-.
  • heterocyclyl refers to an optionally substituted, fully saturated or unsaturated, aromatic or nonaromatic cyclic group, e.g., which is a 4- to 7-membered monocyclic, 7- to 12-membered bicyclic or 10- to 15-membered tricyclic ring system, which has at least one heteroatom in at least one carbon atom-containing ring.
  • Each ring of the heterocyclic group containing a heteroatom may have 1 , 2 or 3 heteroatoms selected from nitrogen atoms, oxygen atoms and sulfur atoms, where the nitrogen and sulfur heteroatoms may also optionally be oxidized.
  • the heterocyclic group may be attached at a heteroatom or a carbon atom.
  • Exemplary monocyclic heterocyclic groups include pyrrolidinyl, pyrrolyl, pyrazolyl, oxetanyl, pyrazolinyl, imidazolyl, imidazolinyl, imidazolidinyl, triazolyl, oxazolyl, oxazolidinyl, isoxazolinyl, isoxazolyl, thiazolyl, thiadiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, furyl, tetrahydrofuryl, thienyl, oxadiazolyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolodinyl, 2-oxoazepinyl, azepinyl, 4-piperidonyl, pyridinyl, pyrazinyl, pyrimidinyl
  • bicyclic heterocyclic groups include indolyl, dihydroidolyl, benzothiazolyl, 4,5,6,7- tetrahydro-benzothiazolyl, benzoxazinyl, benzoxazolyl, benzothienyl, benzothiazinyl, thiazolo[5,4-b]pyridinyl, thiazolo[5,4-d]pyrimidinyl, oxazolo[5,4-b]pyridinyl, 6,7-dihydro-4H- thiopyrano[4,3-d]thiazolyl, 6,7-dihydro-4H-pyrano[4,3-d]thiazolyl, 4,5,6,7-tetrahydro- thiazolo[5,4-c]pyridinyl, 4,5,6,7-tetrahydro-pyrazolo[1 ,5-a]pyridinyl, 5,6,7,8-tetrahydro
  • Exemplary tricyclic heterocyclic groups include carbazolyl, dibenzoazepinyl, dithienoazepinyl, benzindolyl, phenanthrolinyl, acridinyl, phenanthridinyl, phenoxazinyl, phenothiazinyl, xanthenyl, carbolinyl and the like.
  • heterocyclyl includes substituted heterocyclic groups.
  • Substituted heterocyclic groups refer to heterocyclic groups substituted with 1 , 2 or 3 substituents selected from the group consisting of the following:
  • aryl optionally substituted with alkyl, cycloalkyl, alkoxy, hydroxyl, amino, acylamino, alkylamino, dialkylamino or halo.
  • heterocyclooxy denotes a heterocyclic group bonded through an oxygen bridge.
  • heteroaryl refers to an aromatic heterocycle, e.g., monocyclic or bicyclic aryl, such as pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, furyl, thienyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolyl, benzothiazolyl, benzoxazolyl, benzothienyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzofuryl and the like, optionally substituted by, e.g., lower alkyl, lower alkoxy or halo.
  • heteroarylsulfonyl refers to heteroaryl-S(O) 2 -.
  • heteroaroyl refers to heteroaryl-C(O)-.
  • heteroaryloxycarbonyl refers to heteroaryl-O-C(O)-.
  • heteroaroylamino refers to heteroaryl-C(O)NH-.
  • heteroaryl refers to a heteroaryl group bonded through an alkyl group.
  • heteroaralkanoyl refers to heteroaralkyl-C(O)-.
  • heteroaralkanoylamino refers to heteroaralkyl-C ⁇ O)NH-.
  • acyl refers to alkanoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl and the like.
  • acylamino refers to alkanoylamino, aroylamino, heteroaroylamino, aralkanoylamino, heteroaralkanoylamino and the like.
  • esterified carboxy refers to optionally substituted alkoxycarbonyl, cycloalkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, heterocyclooxycarbonyl and the like.
  • salts of the compounds of the present invention refer to salts formed with acids, namely acid addition salts, such as of mineral acids, organic carboxylic acids and organic sulfonic acids, e.g., hydrochloric acid, maleic acid and methanesulfonic acid, respectively.
  • salts of the compounds of the invention refer to salts formed with bases, namely cationic salts, such as alkali and alkaline earth metal salts, e.g., sodium, lithium, potassium, calcium and magnesium, as well as ammonium salts, e.g., ammonium, trimethylammonium, diethylammonium and tris(hydroxymethyl)-methyl- ammonium salts and salts with amino acids provided an acidic group constitutes part of the structure.
  • bases namely cationic salts, such as alkali and alkaline earth metal salts, e.g., sodium, lithium, potassium, calcium and magnesium
  • ammonium salts e.g., ammonium, trimethylammonium, diethylammonium and tris(hydroxymethyl)-methyl- ammonium salts and salts with amino acids provided an acidic group constitutes part of the structure.
  • the present invention provides certain sulfonamide derivatives of formula (I), pharmaceutical compositions containing them, methods for preparing said compounds, and methods of treating glucokinase mediated conditions by administration of a therapeutically effective amount of a compound of the present invention, or a pharmaceutical composition thereof.
  • R 3 is cyclopentyl
  • R 4 is hydrogen
  • R 6 is free or esterified carboxy
  • R 6 is free or esterified carboxy
  • R 6 is free or esterified carboxy
  • R 6 is free or esterified carboxy
  • R 6 is free or esterified carboxy
  • R 6 is free or esterified carboxy
  • R 6 is free or esterified carboxy
  • R 6 is free or esterified carboxy
  • R 6 is free or esterified carboxy
  • an enantiomer thereof or an enantiomeric mixture thereof
  • pharmaceutically acceptable salt thereof are pharmaceutically acceptable salt thereof.
  • R 9 is hydrogen, optionally substituted lower alkyl, lower alkenyl, cycloalkyl, aryl or aralkyl;
  • n is an integer from 1 to 3; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
  • n is an integer from 1 to 3;
  • R 9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
  • n is an integer from 1 to 3;
  • R 1 is hydrogen, halogen, cyano, trifluoromethyl, alkoxy, alkylthio or carboxy;
  • R 2 is absent; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
  • n is an integer from 1 to 3;
  • R 1 is hydrogen, halogen, cyanb, trifluoromethyl, alkoxy, alkylthio or carboxy; R 2 is absent;
  • R 5 is (C 3-7 )cycloalkyl or (C 1-4 )alkyl substituted by (C 1-6 )alkoxy, (C 3-7 )cycloalkyl, (C 6-1 o)aryl or (C 5-6 )heterocyclyl; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
  • R 5 is (C 3-7 )cycloalkyl or (C 1-4 )alkyl substituted by (C 1-6 )alkoxy, (C 3 . 7 )cycloalkyl, (C 6- i 0 )aryl or (C 5 - 6 )heterocyclyl;
  • R 9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
  • R 5 is (C 3 . 7 )cycloalkyl or (C ⁇ alkyl substituted by (Ci -6 )alkoxy, (C 3-7 )cycloalkyl, (C 6- io)aryl or (C 5 . 6 )heterocyclyl in which (Ci -4 )alkyl is methyl or ethyl, (C 1-6 )alkoxy is methoxy or ethoxy; and (C 6- io)aryl is optionally substituted phenyl;
  • R 9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
  • R 9 is hydrogen, optionally substituted lower alkyl, lower alkenyl, cycloalkyl, aryl or aralkyl;
  • n is an integer from 1 to 3; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
  • n is an integer from 1 to 3;
  • R 9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
  • n is an integer from 1 to 3;
  • R 1 is hydrogen, halogen, cyano, trifluoromethyl, alkoxy, alkylthio or carboxy;
  • R 2 is absent; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
  • n is an integer from 1 to 3;
  • R 1 is hydrogen, halogen, cyano, trifluoromethyl, alkoxy, alkylthio or carboxy; R 2 is absent;
  • R 5 is (C 3 . 7 )cycloalkyl or (C 1-4 )alkyl substituted by (C 1-6 )alkoxy, (C 3-7 )cycloalkyl, (C 6-10 )aryl or (C 5 . 6 )heterocyclyl; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
  • R 5 is (C 3 . 7 )cycloalkyl or (C 1-4 )alkyl substituted by (C 1-6 )alkoxy, (C 3-7 )cycloalkyl, (C 6- io)aryl or (C 5 . 6 )heterocyclyl;
  • R 9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
  • R 5 is (C 3-7 )cycloalkyl or (C-
  • R 9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
  • the compounds of the invention depending on the nature of the substituents possess one or more asymmetric centers.
  • the resulting diastereoisomers, optical isomers, i.e., enantiomers, and geometric isomers, and mixtures thereof, are encompassed by the instant invention.
  • Preferred are the compounds of the present invention wherein the substituent at the carbon atom adjacent to the amide group attains the R-configuration.
  • Particular embodiments of the invention are:
  • FV and R 2 ' represents R 1 and R 2 , respectively, as defined herein above, or R 1 ' and R 2 1 are groups convertible to R 1 and R 2 , respectively, with an activated derivative of a carboxylic acid of the formula
  • R 3 , R 4 and n have meanings as defined herein, and R 5 ' and R 6 ' represents R 5 and R 6 , respectively, as defined herein above, or R 5 ' and R 6 ' are groups convertible to R 5 and R 6 , respectively, to afford a compound of the formula wherein R 1 ', R 2 ', R 3 , R 4 , n, R 5 ' and R 6 ' have meanings as defined for formulae (II) and (III).
  • an activated derivative of a carboxylic acid e.g., those corresponding to carboxylic acids of formula (III)
  • coupling agents such as 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride
  • Mixed anhydrides are preferably such from pivalic acid, or lower alkyl hemiesters of carbonic acids, such as ethyl or isobutyl analogs.
  • Activated esters include, for example, succinimido, phthalimido or 4-nitrophenyl esters.
  • reaction of an activated derivative of a carboxylic acid, e.g., those corresponding to carboxylic acids of formula (III), with an amine, e.g., those of formula (II), may be carried out in the presence of a base, such as pyridine, triethylamine (TEA), diisopropylethylamine (DIEA) or N-methylmorpholine (NMM) in an inert organic solvent, such as dichloromethane (DCM), N,N-dimethylformamide (DMF) or tetrahydrofuran (THF), or a mixture of solvents thereof.
  • a base such as pyridine, triethylamine (TEA), diisopropylethylamine (DIEA) or N-methylmorpholine (NMM)
  • an inert organic solvent such as dichloromethane (DCM), N,N-dimethylformamide (DMF) or tetrahydrofuran (THF), or a
  • Carboxylic acids of formula (III) may be converted to their activated derivatives using methods described herein or according to methods generally known in the art, e.g., a carboxylic acid of formula (III) may be treated with a chlorinating agent, such as thionyl chloride or oxalyl chloride, to afford a corresponding acid chloride thereof, or by the treatment of a coupling agent such as EDCI or HOBt, or a mixture of coupling agents thereof.
  • a chlorinating agent such as thionyl chloride or oxalyl chloride
  • Amines of formula (II) and carboxylic acids of formula (III) are known, or if they are novel they may be prepared using methods well known in the art or as described herein in the illustrative Examples, or modifications thereof.
  • compounds of formula (III) may be prepared by treating an ester of the formula wherein R 4 has a meaning as defined herein above, and R is lower alkyl, preferably, methyl or ethyl, with chlorosulfonic acid to afford a compound of the formula
  • R 4 and R have meanings as defined herein above, optionally in the presence of an intrinsic organic solvent.
  • the reaction is carried out without an intrinsic organic solvent.
  • a compound of formula (V) may then be treated with an amine of the formula
  • n, R 5 ' and R 6 ' have meanings as defined herein above, in the presence of a base, such as pyridine, TEA, DIEA or NMM, in an inert organic solvent, such as DCM, DMF or THF, or a mixture of solvents thereof, to afford a compound of the formula
  • reaction is conducted at a temperature ranging from about -4 0 C to room temperature (RT), more preferably, the reaction temperature is about O 0 C.
  • Amines of formula (Vl) are known, or if they are novel they may be prepared using methods well known in the art or as described herein in the illustrative Examples.
  • a resulting compound of formula (VII) may then be treated with a base, such as sodium hydride, lithium diisopropylamide (LDA) or lithium bis(trimethylsilyl)amide (LHMDS), preferably LDA, followed by addition of an alkylating agent of the formula R 3 -(CH 2 )-Lg (VIII) wherein R 3 has a meaning as defined herein above, and Lg represents a leaving group, such as chloride, bromide, iodide, mesylate, tosylate or triflate, preferably iodide ot triflate, to afford a compound of the formula
  • the alkylation step is preferably conducted in a polar organic solvent, such as THF, DMF, N-methylpyrrolidone (NMP), 1 ,3-dimethyl-3,4,5,6-tetrahydro-2('//-/)-pyridone (DMPU) or 1 ,3- dimethyl-3,4,5,6-tetrahydro-2(7/-/)-pyrimidinone (DMTP), or in a mixture of solvents thereof.
  • a polar organic solvent such as THF, DMF, N-methylpyrrolidone (NMP), 1 ,3-dimethyl-3,4,5,6-tetrahydro-2('//-/)-pyridone (DMPU) or 1 ,3- dimethyl-3,4,5,6-tetrahydro-2(7/-/)-pyrimidinone (DMTP), or in a mixture of solvents thereof.
  • a resulting compound of formula (IX) may then be hydrolyzed, e.g., in the presence of an aqueous base such as sodium, lithium or potassium hydroxide and an organic solvent such as THF or lower alcohol, preferably, methanol or ethanol, to afford a carboxylic acid of formula (III) wherein R 3 , R 4 , n, R 5 ' and R 6 ' have meanings as defined herein above.
  • an aqueous base such as sodium, lithium or potassium hydroxide
  • organic solvent such as THF or lower alcohol, preferably, methanol or ethanol
  • a carboxylic acid of formula (III) may then be coupled with an amine of formula (II), or an acid addition salt thereof, under reaction conditions as described herein above to afford a compound of formula (I 1 ) wherein R 1 ', R 2 ', R 3 , R 4 , n, R 5 ' and R 6 ' have meanings as defined herein above, e.g., via conversion of the acid to the corresponding acid chloride or in the presence of a coupling agent such as EDCI, HOBt or PyBOP, or a mixture of coupling agents thereof.
  • a coupling agent such as EDCI, HOBt or PyBOP, or a mixture of coupling agents thereof.
  • Compounds of formula (I) may be obtained by reacting a compound of the formula wherein R 3 and R 4 have meanings as defined herein above, and R 1 ' and R 2 ' represents Ri and R 2 , respectively, as defined herein above, or R 1 ' and R 2 ' are a groups convertible to Ri and R 2 , respectively, with an amine of the formula
  • R 1 ', R 2 ', R 3 , R 4 , n, R 5 ' and R 6 ' have meanings as defined herein above.
  • R 4 and R have meanings as defined herein above, with a base, such as sodium hydride, LDA or LHMDS, preferably LDA, followed by addition of an alkylating agent of the formula
  • the alkylation step is preferably conducted in a polar organic solvent, such as THF, DMF, NMP, DMPU or DMTP, or in a mixture of solvents thereof.
  • a resulting compound of formula (XII) may then be hydrolyzed, e.g., in the presence of an aqueous base, such as sodium, lithium or potassium hydroxide and an organic solvent such as THF or lower alcohol, preferably, methanol or ethanol, to afford a carboxylic acid of the formula
  • R 3 and R 4 have meanings as defined herein above.
  • a carboxylic acid of formula (XIII) may then be coupled with an amine of formula (II) under reaction conditions as described herein above to afford a compound of the formula
  • R 1 ', R2', R 3 and R 4 have meanings as defined herein above, e.g., via conversion of the carboxylic acid to the corresponding acid chloride, or in the presence of a coupling agent, such as EDCI, HOBt or PyBOP, or a mixture of coupling agents thereof.
  • a coupling agent such as EDCI, HOBt or PyBOP, or a mixture of coupling agents thereof.
  • a resulting compound of formula (XIV) may then be converted to a sulfonyl chloride derivative of formula (X) wherein R 1 ', R 2 ', R 3 and R 4 have meanings as defined herein above, by reduction of the nitro group to the amino group, e.g., using iron powder in the presence of a mixture of acetic acid and a lower alcohol, such as ethanol, followed by diazotization reaction and subsequent treatment with, e.g., sulfur dioxide in the presence of copper(ll) chloride and acetic acid.
  • a resulting sulfonyl chloride derivative of formula (X) may then be treated with an amine of formula (Vl), or an acid addition salt thereof, wherein n, R 5 ' and R 6 ' have meanings as defined herein above, under reaction conditions described herein above to afford a compound of formula (I 1 ) wherein R 1 ', R 2 ', R 3 , R 4 , n, R 5 ' and R 6 ' have meanings as defined herein above.
  • protecting groups are to protect the functional groups from undesired reactions with reaction components under the conditions used for carrying out a desired chemical transformation.
  • the need and choice of protecting groups for a particular reaction is known to those skilled in the art and depends on the nature of the functional group to be protected (hydroxyl group, amino group, etc.), the structure and stability of the molecule of which the substituent is a part and the reaction conditions.
  • the invention further includes any variant of the present processes, in which an intermediate product obtainable at any stage thereof is used as starting material and the remaining steps are carried out, or in which the starting materials are formed in situ under the reaction conditions, or in which the reaction components are used in the form of their salts or optically pure antipodes.
  • the invention also relates to any novel starting materials, intermediates and processes for their manufacture.
  • the new compounds may be in the form of one of the possible isomers or mixtures thereof, for example, as substantially pure geometric (cis or trans) isomers, diastereomers, optical isomers (enantiomers, antipodes), racemates or mixtures thereof.
  • the aforesaid possible isomers or mixtures thereof are within the purview of this invention.
  • Any resulting mixtures of isomers can be separated on the basis of the physicochemical differences of the constituents, into the pure geometric or optical isomers, diastereomers, racemates, for example, by chromatography and/or fractional crystallization.
  • any resulting racemates of final products or intermediate e.g., acids of formulae (III) and (XIII)
  • the compounds of the instant invention having a basic moiety may be resolved into their optical isomers, e.g., by fractional crystallization of a salt formed with an optically active acid, e.g., tartaric acid, dibenzoyl tartaric acid, diacetyl tartaric acid, di-0,0'-p-toluoyl tartaric acid, mandelic acid, malic acid or camphor-10-sulfonic acid. Racemic products can also be resolved by chiral chromatography, e.g., high pressure liquid chromatography (HPLC) using a chiral adsorbent.
  • HPLC high pressure liquid chromatography
  • optically pure isomers of compounds of the present invention may be obtained by employing chiral reagents.
  • an optical isomer, preferably the R isomer, of compounds of formula (X) may be prepared employing chiral auxiliaries, e.g., the Evans auxiliary, as illustrated herein in the Examples.
  • compounds of the invention are either obtained in the free form, or in a salt form thereof, preferably, in a pharmaceutically acceptable salt form thereof, or as a prodrug derivative thereof.
  • salts may be converted into salts with pharmaceutically acceptable bases.
  • Such salts include alkali metal salts, like sodium, lithium and potassium salts; alkaline earth metal salts, like calcium and magnesium salts; ammonium salts with organic bases, e.g., trimethylamine salts, diethylamine salts, tris(hydroxymethyl)methylamine salts, dicyclohexylamine salts and N-methyl-D-glucamine salts; salts with amino acids like arginine, lysine and the like. Salts may be formed using conventional methods, advantageously in the presence of an ethereal or alcoholic solvent, such as a lower alkanol.
  • the salts may be precipitated with ethers, e.g., diethyl ether. Resulting salts may be converted into the free compounds by treatment with acids. These or other salts can also be used for purification of the compounds obtained.
  • Compounds of the invention may be converted into acid addition salts, especially pharmaceutically acceptable salts. These are formed, e.g., with inorganic acids, such as mineral acids, e.g., sulfuric acid, phosphoric or hydrohalic acid, or with organic carboxylic acids, such as (Ci- 4 )alkanecarboxylic acids which, e.g., are unsubstituted or substituted by halogen, e.g., acetic acid, such as saturated or unsaturated dicarboxylic acids, e.g., oxalic, succinic, maleic or fumaric acid, such as hydroxycarboxylic acids, e.g., glycolic, lactic, malic, tartaric or citric acid, such as amino acids, e.g., aspartic or glutamic acid, or with organic sulfonic acids, such as (Cr 4 )alkylsulfonic acids, e.g., methanesulf
  • Prodrug derivatives of any compound of the invention are derivatives of said compounds which following administration release the parent compound in vivo via some chemical or physiological process, e.g., a prodrug on being brought to the physiological pH or through enzyme action is converted to the parent compound.
  • exemplary prodrug derivatives are, e.g., esters of free carboxylic acids and S-acyl and O-acyl derivatives of thiols, alcohols or phenols, wherein acyl has a meaning as defined herein.
  • Preferred are pharmaceutically acceptable ester derivatives convertible by solvolysis under physiological conditions to the parent carboxylic acid.
  • ester derivatives include, but are not limited to, lower alkyl esters, cycloalkyl esters, lower alkenyl esters, benzyl esters, mono- or di-substituted lower alkyl esters, such as the ⁇ -(amino, mono- or di-lower alkylamino, carboxy, lower alkoxycarbonyl)-lower alkyl esters, the ⁇ -(lower alkanoyloxy, lower alkoxycarbonyl or di-lower alkylaminocarbonyl)-lower alkyl esters, such as the pivaloyloxymethyl ester and others conventionally used in the art, e.g., a compound of formula (IA) wherein R 9 is optionally substituted lower alkyl as described herein above, lower alkenyl, cycloalkyl, aryl or aralkyl, is easily convertible to a compound of formula (IA) wherein R 9 is hydrogen.
  • prodrug derivatives In view of the close relationship between the free compounds, the prodrug derivatives and the compounds in the form of their salts, whenever a compound is referred to in this context, a prodrug derivative and a corresponding salt is also intended, provided such is possible or appropriate under the circumstances.
  • the compounds, including their salts, can also be obtained in the form of their hydrates, or include other solvents used for their crystallization.
  • the compounds of the present invention may be employed for the treatment of conditions mediated by glucokinase activity. Such compounds may thus be employed therapeutically for the treatment of impaired glucose tolerance, type 2 diabetes and obesity.
  • the present invention further provides pharmaceutical compositions comprising a therapeutically effective amount of a pharmacologically active compound of the instant invention, alone or in combination with one or more pharmaceutically acceptable carriers.
  • compositions according to the invention are those suitable for enteral, such as oral or rectal; transdermal and parenteral administration to mammals, including man, for the treatment of conditions mediated by glucokinase activity.
  • enteral such as oral or rectal
  • transdermal and parenteral administration to mammals, including man, for the treatment of conditions mediated by glucokinase activity.
  • Such conditions include impaired glucose tolerance, type 2 diabetes and obesity.
  • the pharmacologically active compounds of the invention may be employed in the manufacture of pharmaceutical compositions comprising an effective amount thereof in conjunction or admixture with excipients or carriers suitable for either enteral or parenteral application.
  • Injectable compositions are preferably aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions.
  • compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. In addition, they may also contain other therapeutically valuable substances.
  • adjuvants such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers.
  • Said compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1-75%, preferably about 1-50%, of the active ingredient.
  • Suitable formulations for transdermal application include a therapeutically effective amount' of a compound of the invention with carrier.
  • Advantageous carriers include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host.
  • transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound of the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • the present invention provides pharmaceutical compositions as described above for the treatment of conditions mediated by glucokinase activity, preferably, impaired glucose tolerance, type 2 diabetes and obesity.
  • compositions may contain a therapeutically effective amount of a compound of the invention as defined above, either alone or in a combination with another therapeutic agent, e.g., each at an effective therapeutic dose as reported in the art.
  • therapeutic agents include: a) anti-diabetic agents, such as insulin, insulin derivatives and mimetics; insulin secretagogues such as the sulfonylureas, e.g., Glipizide, glyburide and Amaryl; insulinotropic sulfonylurea receptor ligands such as meglitinides, e.g., nateglinide and repaglinide; thiazolidone derivatives such as glitazones, e.g., pioglitazone and rosiglitazone; protein tyrosine phosphatase-1 B (PTP-1 B) inhibitors such as PTP-112; GSK3 (glycogen synthase kinase-3) inhibitors such as SB
  • DPPIV dipeptidyl peptidase IV
  • DPPIV dipeptidyl peptidase IV
  • SCD-1 stearoyl-CoA desaturase-1 inhibitors
  • DGAT1 and DGAT2 diacylglycerol acyltransferase 1 and 2) inhibitors
  • ACC2 acetyl CoA carboxylase 2 inhibitors
  • breakers of AGE advanced glycation end products
  • b) anti-dyslipidemic agents such as 3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase inhibitors, e.g., lovastatin, pitavastatin, simvastatin, pravastatin, cerivastatin, mevastatin, velostatin, fluvastatin, dalvastatin, atorvastatin,
  • HMG-CoA 3-hydroxy-3-methyl-glutaryl coenzyme A reductase inhibitor
  • a compound of the present invention may be administered either simultaneously, before or after the other active ingredient, either separately by the same or different route of administration or together in the same pharmaceutical formulation.
  • compositions comprising a therapeutically effective amount of a compound of the invention in combination with a therapeutically effective amount of another therapeutic agent, preferably selected from antidiabetics, hypolipidemic agents, anti-obesity agents or anti-hypertensive agents, most preferably from antidiabetics or hypolipidemic agents as described above.
  • another therapeutic agent preferably selected from antidiabetics, hypolipidemic agents, anti-obesity agents or anti-hypertensive agents, most preferably from antidiabetics or hypolipidemic agents as described above.
  • the present invention further relates to pharmaceutical compositions as described above for use as a medicament.
  • the present invention further relates to use of pharmaceutical compositions or combinations as described above for the preparation of a medicament for the treatment of conditions mediated by glucokinase activity, preferably, impaired glucose tolerance, type 2 diabetes and obesity.
  • the present invention also relates to a compound of formula (I) for use as a medicament; to the use of a compound of formula (I) for the preparation of a pharmaceutical composition for the prevention and/or treatment of conditions mediated by glucokinase activity, and to a pharmaceutical composition for use in conditions mediated by glucokinase activity comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable diluent or carrier therefore.
  • the present invention further provides a method for the prevention and/or treatment of conditions mediated by glucokinase activity, which comprises administering a therapeutically effective amount of a compound of the present invention.
  • a unit dosage for a mammal of about 50-70 kg may contain between about 1 mg and 1000 mg, advantageously between about 5-500 mg of the active ingredient.
  • the therapeutically effective dosage of active compound is dependent on the species of warm-blooded animal (mammal), the body weight, age and individual condition, on the form of administration, and on the compound involved.
  • the present invention also provides a therapeutic combination, e.g., a kit, kit of parts, e.g., for use in any method as defined herein, comprising a compound of formula (I), or a pharmaceuticallyacceptable salt thereof, to be used concomitantly or in sequence with at least one pharmaceutical composition comprising at least another therapeutic agent, preferably selected from anti-diabetic agents, hypolipidemic agents, anti-obesity agents and anti-hypertensive agents, or a pharmaceutically acceptable salt thereof.
  • the kit may comprise instructions for its administration.
  • kits of parts comprising: (i) a pharmaceutical composition of the invention; and (ii) a pharmaceutical composition comprising a compound selected from an anti-diabetic, a hypolipidemic agent, an anti-obesity agent and an antihypertensive agent, or a pharmaceutically acceptable salt thereof, in the form of two separate units of the components (i) to (ii).
  • the present invention provides a method as defined above comprising coadministration, e.g., concomitantly or in sequence, of a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second drug substance, said second drug substance being an anti-diabetic, a hypolipidemic agent, an anti-obesity agent or an anti-hypertensive agent, e.g., as indicated above.
  • a compound of the invention is administered to a mammal in need thereof.
  • a compound of the invention is used for the treatment of a disease which responds to modulation of the glucokinase activity.
  • the condition associated with glucokinase activity is selected from impaired glucose tolerance, type 2 diabetes and obesity.
  • the present invention provides a method or use which comprises administering a compound of formula (I) in combination with a therapeutically effective amount of an antidiabetic agent, a hypolipidemic agent, an anti-obesity agent or an anti-hypertensive agent.
  • the present invention provides a method or use which comprises administering a compound of formula (I) in the form of a pharmaceutical composition as described herein.
  • treatment embraces all the different forms or modes of treatment as known to those of the pertinent art and in particular includes preventive, curative, delay of progression and palliative treatment.
  • the above-cited properties are demonstrable in vitro and in vivo tests using advantageously mammals, e.g., mice, rats, dogs, monkeys or isolated organs, tissues and preparations thereof.
  • Said compounds can be applied in vitro in the form of solutions, e.g., preferably aqueous solutions, and in vivo either enterally, parenterally, advantageously intravenously, e.g., as a suspension or in aqueous solution.
  • the dosage in vitro may range between about 10 "2 molar and 10 "9 molar concentrations.
  • a therapeutically effective amount in vivo may range depending on the route of administration, between about 0.1 mg/kg and 1000 mg/kg, preferably between about 1 mg/kg and 100 mg/kg.
  • the activity of compounds according to the invention may be assessed by the following methods or methods well-described in the art:
  • the glucokinase activation in vitro may be determined by measuring the activation of recombinant GST-GK by a compound of the present invention in the absence or the presence of GKRP, a 68,000 Da protein inhibitor of GK.
  • formation of glucose-6-phosphate is coupled directly to the formation of thio-NADH.
  • GST-GK catalyzes the reaction of glucose and Mg-ATP to produce glucose-6-phosphate and ADP.
  • Glucose-6- phosphate dehydrogenase (G6PDH) reduces thionicotinamide (thio-NAD) to thio-NADH.
  • the assay measures the formation of thio-NADH at 405 nM.
  • the basic GK assay components are as follows: 25 mM HEPES (pH 7.1), 25 mM KCI, 2.5 mM MgCI 2 , 1 mM ATP (Sigma A-5394), 1 mM DTT, 1 mM thio-NAD (Sigma T-7375), 80 units/mL G6PDH (Sigma G-5885), 10 mM glucose and 110 nM GST-GK.
  • 20 ⁇ M Fructose-6-phosphate (F-6-P) and 370 nM recombinant GKRP are added to these assay components.
  • Fructose-1 -phosphate (F-1-P) at 1 ⁇ M is used as a control in the GK/GKRP assay.
  • F-1-P reverses inhibition of GST-GK by GKRP.
  • the assay is done in standard, 96-well, round-bottom plates (Corning) and the total assay volume is 25 ⁇ l_.
  • Test compounds are serially diluted into 100% DMSO and 0.5 ⁇ l_ of diluted compound in 100% DMSO is added to the assay plate.
  • Assay reagents (24.5 ⁇ l_) are added using a Zymark robotic platform.
  • Buffer containing HEPES, MgCI 2 , KCI, thio-NAD, G6PDH, glucose and GST-GK, are added (5 ⁇ L) using the Zymark 8-channel hand pipet.
  • GKRP and F-6-P are also included.
  • the reaction is then initiated by adding 19.5 ⁇ L of buffer containing HEPES, MgCI 2 , KCI, DTT and ATP using the Zymark Reagent Addition Station/Reagent Addition Module.
  • the plates are read kinetically over 10 min at 25°C using a SpectraMax Plus microplate spectrophotometer (Molecular Devices, Sunnyvale, CA) to monitor the increase in optical density at 405 nm.
  • the GK activity in wells containing test compounds is compared with activity in DMSO control wells.
  • the concentration of compound that produces a 50% increase in the activity of GK is calculated and expressed as EC 5 O-
  • a majority of the compounds described in the Examples had an EC 50 value less than or equal to 200 ⁇ M and preferably less than 50 ⁇ M. Most preferable are compounds with EC 50 less than 2 ⁇ M which exhibited at least a 2-fold increase in % GK activation versus control.
  • glucokinase activation in rat hepatocytes may be determined as follows: Hepatocytes are isolated by collagenase perfusion of the livers of overnight-fasted male Harlen Sprague-Dawley rats (Charles River Laboratories, Raleigh, NC) as previously described (see Berry et al., J. Cell Biol., Vol. 43, pp. 506-520 (1969)). The cells are washed three times each with 100 ml. of glucose-free Dulbecco's Modified Eagle medium (DMEM, Gibco BRL) containing 5% fetal bovine serum (FBS) and then suspended in glucose-free DMEM/5% FBS.
  • DMEM glucose-free Dulbecco's Modified Eagle medium
  • FBS fetal bovine serum
  • Cells are plated in collagen coated 24-well plates (Becton Dickinson) at a density of 3 x 10 5 cells/well in 1 mL of William's Medium E (Sigma) supplemented with 5% FBS, and incubated at 37°C in 5% CO 2 /95% air. After cell attachment ( ⁇ 4 h), the medium is replaced with serum-free DMEM containing 5 mM glucose and 10 nM dexamethasone (Sigma), and cells are cultured further for 16-20 h prior to use.
  • DMEM serum-free DMEM containing 5 mM glucose and 10 nM dexamethasone
  • the rate of glucose phosphorylation is determined by the release of 3 H 2 O from [2- 3 H]glucose.
  • the medium from the cultured hepatocytes is removed, and the cells are pre-incubated in 150 ⁇ L of fresh serum-free DMEM containing 5 mM glucose and compound (1 , 10 and 30 ⁇ M) or DMSO for 3 h at 37 0 C.
  • the final concentration of DMSO is 0.2%.
  • the medium is then removed and 150 ⁇ L of a fresh mixture of DMEM/5 mM glucose containing compound or DMSO, and 1 ⁇ Ci of [2- 3 H]glucose (NEN) is added.
  • cells are pre-incubated in serum-free DMEM/5 mM glucose medium containing DMSO for 3 h and then are incubated for 1 h in labeled glucose medium containing 0.5 mM fructose/DMSO (precursor of F-1-P, AnalaR ® from BDH). All conditions are tested in quadruplicate where one well per plate received 200 ⁇ L of the appropriate medium plus labeled glucose (instead of 150 ⁇ L) of which 50 ⁇ L is immediately removed and placed in a 1.2 mL microfuge tube (Costar) containing 10 ⁇ L of 1 N HCI. This sample is used as a 0-minute time point for determining background 3 H 2 O release (exchange values). Following the addition of the labeled glucose media, hepatocytes are incubated at 37°C on a slow moving rocker for 1 h.
  • labeled glucose medium containing 0.5 mM fructose/DMSO (precursor of F-1-P, AnalaR ® from B
  • the culture medium On termination of the incubation, 50 ⁇ L of the culture medium is collected into microfuge tubes containing 10 ⁇ L of 1 N HCI, and determination of 3 H 2 O.
  • the tubes are left uncapped and each is placed inside a 20 mL glass scintillation vial (Wheaton) containing 1.5 mL of deionized water.
  • the vials are capped tightly and incubated at 37°C in a dry incubator for 2 days ( 3 H 2 O from the reaction mixture will equilibrate with the water in the vial).
  • a standard curve is generated using [ 3 H]H 2 O (NEN) to correct for exchange.
  • the glucokinase activation in vivo may be determined as follows:
  • mice Male C57BL mice (Jackson Lab, Bar Harbor, ME) are housed 2 per cage in a reversed light cycle room (light on from 8:00 p.m. to 8:00 a.m.) and given access to food and water ad libitum.
  • DIO high fat diet
  • the mice are given a high fat diet (D12492 with 60% caloric intake from fat, Research Diets, New Brunswick, NJ) from 4 weeks of age and maintained on the diet before being used.
  • the DIO mice are used at 25 weeks of age.
  • animals On the day of the study, animals are fasted at 7:30 a.m.
  • Body weight measurement and basal blood sample collection are conducted at 10:00 a.m. Plasma glucose values are then determined.
  • animals are dosed with vehicle (water) or compound in vehicle with a dose volume of 5 ml/kg.
  • the test compound is given at 3, 10, 30, or 100 mg/kg.
  • a blood sample (at 0 min) is taken followed by an oral glucose tolerance test (OGTT) at 1 g/kg (20% glucose in water) and a dose volume of 5 ml/kg. Blood samples are collected at 30, 60 and 120 min following the glucose administration. The animals are refed after the OGTT. Blood samples are taken via tail bleeding.
  • OGTT oral glucose tolerance test
  • Plasma glucose concentrations are determined using a glucose meter (Ascensia Elite, Bayer Corp., Mishawaka, IN). Blood samples are collected in tubes (Microvette CB300, Aktiengesellschaft & Co., Numbrecht, Germany) which contain EDTA (ethylene diaminetetraacetic acid) to prevent blood clotting. After blood sample collection, the tubes are kept on ice before being centrifuged. Plasma portion of the blood samples is obtained by centrifugation at 10,000 x g for 10 min at 4 0 C and then stored at -80 0 C. Plasma insulin levels are determined by Luminex assay using Mouse Endocrine Lincop/ex kit (Linco Research, Inc., St. Charles, MO). Data are reported as means ⁇ SEM.
  • the compound of Example 1 demonstrates an EC 50 of about 271 nM in the in vitro assay measuring the activation of recombinant GST-GK.
  • Oral (po) dosing of a compound of Example 1 provides statistically significant glucose lowering with reductions in glucose of 38-53% at different timepoints during the oral glucose tolerance test.
  • Triflurormethanesulfonic anhydride (6.72 mL, 39.94 mmol) is dissolved in DCM (40 ml_) and cooled to -4O 0 C. To this is added dropwise cyclopentanemethanol (4.00 g, 39.94 mmol) dissolved in DCM (10 mL) and anhydrous pyridine (3.23 mL, 39.94 mmol). The reaction is stirred at -4O 0 C for 45 min. The reaction is diluted with hexanes (30 mL) and run through a plug of silica using hexanes (300 mL) as the eluent. Concentration without heating gives trifluoromethanesulfonic acid cyclopentylmethyl ester as a clear, colorless oil that is used immediately.
  • reaction is allowed to stir and warm to RT. After one hour, the reaction is concentrated to remove the solvent, resulting in a yellow oil. This is dissolved in ethyl acetate (300 mL) and washed with water (2 x 200 mL). The organic solution is dried over anhydrous sodium sulfate, filtered, and concentrated to a yellow oil.
  • Lithium hydroxide monohydrate (1.28 g , 30.6 mmol) is suspended in THF (50 mL) and cooled to O 0 C. Hydrogen peroxide (50 wt% in water, 2.81 mL, 91.79 mmol) is added dropwise. Reaction is stirred at O 0 C for 15 min and the title C compound, (4R,5S)-3-((R)-3- cyclopentyl-2-phenyl-propionyl)-4-methyl-5-phenyl-oxazolidin-2-one (3.85 g, 10.2 mmol) is added in five separate portions, allowing portions to dissolve completely between additions. The reaction is allowed to stir and warm to RT overnight.
  • Extracts are combined, dried over anhydrous sodium sulfate, filtered, and concentrated to give 4-[(R)-2-cyclopentyl-1-(5-methoxy-thiazolo[5,4- 6]pyridin-2-ylcarbamoyl)-ethyl] benzenesulfonyl chloride as a yellow foam.
  • Benzylamine (1 g, 9.33 mmol) is dissolved in ethanol (25 ml_), to which tert-butyl acrylate (1.20 g, 9.33 mmol) is added. The reaction is stirred at RT overnight and then concentrated to a crude yellow oil.
  • the organic layer is evaporated to a light orange resin which is dissolved in 2 mL methanol in a 50 mL round bottom flask.
  • Aqueous potassium hydroxide (0.265 mL of 0.5094 M potassium hydroxide solution) is added and stirred for 30 min.
  • reaction mixture is stirred at -78 0 C for 30 min and then cyclopentyl methyl iodide (17.6 g, 78 mmol) in 50 mL of 9:1 THF/DMPU is added.
  • the reaction is stirred while warming slowly to RT overnight.
  • the mixture is poured into 1 L of 1 N HCI and extracted twice with MTBE.
  • the combined MTBE extracts are washed with brine, dried over anhydrous magnesium sulfate, filtered and reduced to an orange oil.
  • the resulting yellow solution is then added to 180 mL of the Green Solution (prepared by bubbling 74 g of sulfur dioxide gas into 740 mL of glacial acetic acid followed by addition of 30 g of CuCb in 35-40 mL water.
  • the resulting mixture is filtered through filter paper to obtain a clear green solution) and the mixture is stirred at RT overnight (the initial black- green solution transforms to a light green solution after 24 h).
  • the resulting mixture is poured onto 500 g of ice and the precipitated solids are collected by filtration, washed with water and then dissolved in ethyl acetate, washed with brine, dried over anhydrous magnesium sulfate, filtered and evaporated to afford a yellow foam.
  • Benzofuran-2-yl-methanol 450 mg, 3.037 mmol is dissolved in THF (100 mL).
  • Triphenylphosphine 956 mg, 3.645 mmol
  • phthalamide 536 mg, 3.645 mmol
  • the reaction is cooled to 0 0 C.
  • Di-tert-butyl azodicarboxylate 839 mg, 3.645 mmol is dissolved in THF (10 mL) and added dropwise. The reaction is allowed to warm to RT overnight and the solvent is removed by rotary evaporation.

Abstract

The present invention provides compounds of the formula (I), which are activators of glucokinase activity and, thus, may be employed as therapeutic agents for the treatment of glucokinase mediated conditions. Accordingly, the compounds of formula (I) may be employed for the prevention and the treatment of impaired glucose tolerance, type 2 diabetes and obesity.

Description

SULFONAMIDE DERIVATIVES AS GLYCOKINASE ACTIVATORS USEFUL IN THE TREATMENT OF TYPE 2 DIABETES
The present invention relates to certain sulfonamide derivatives, pharmaceutical compositions containing them, and to methods of treating glucokinase mediated conditions, in particular, impaired glucose tolerance and type 2 diabetes, by employing such compounds.
Accordingly, the present invention provides compounds of the formula
Figure imgf000002_0001
wherein
Q combined together with the carbon and nitrogen atoms to which it is attached form a 5- to 6-membered monocyclic heteroaromatic ring; or
Q combined together with the carbon and nitrogen atoms to which it is attached form a 9- to 10-membered bicyclic heterocycle;
R1 and R2 are, independently from each other, hydrogen, halogen, cyano, nitro, optionally substituted alkyl, alkoxy, alkylthio, alkylthiono, sulfonyl, free or esterified carboxy, carbamoyl, sulfamoyl, optionally substituted amino, aryl or heterocyclyl; or
R2 is absent;
R3 is (C3.6)cycloalkyl or (C3-6)heterocyclyl;
R4 is hydrogen, halogen, cyano, lower alkyl or lower alkoxy;
R5 is (C3-12)cycloalkyl, (C6.io)aryl, (C3-10)heterocyclyl or (d.6)alkyl optionally substituted by (C1-6)alkoxy, (C1-2)alkoxy-(Ci-4)alkoxy, (C1-6)alkylthio, (C3-7)cycloalkyl, (C3-7)cycloalkoxy, (C3-7)cycloalkylthio, (C6-10)aryl, (C6-i0)aryloxy, (C6-10)arylthio, (C3-1 o)heterocyclyl or (C3-10)heterocyclyloxy;
R6 is free or esterified carboxy, tetrazolyl, cyano or -C(O)NR7R8 in which
R7 and R8 are, independently from each other, hydrogen, optionally substituted alkyl, cycloalkyl, aryl or heterocyclyl; or R7 and R8 combined are alkylene which together with the nitrogen atom to which they are attached form a 4- to 7-membered ring; n is an integer from 1 to 6; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
The compounds of the present invention provide pharmacological agents which are glucokinase activators and, thus, may be employed for the treatment of glucokinase mediated conditions. Accordingly, the compounds of formula (I) may be employed for prevention and treatment of impaired glucose tolerance, type 2 diabetes and obesity.
Listed below are definitions of various terms used to describe the compounds of the present invention. These definitions apply to the terms as they are used throughout the specification unless they are otherwise limited in specific instances either individually or as part of a larger group, , e.g., wherein an attachment point of a certain group is limited to a specific atom within that group, the point of attachment is defined by an arrow at the specific atom.
The term "optionally substituted alkyl" refers to unsubstituted or substituted alkyl groups, i.e., straight- or branched-chain hydrocarbon groups having 1-20 carbon atoms, preferably 1-10 carbon atoms. Exemplary unsubstituted alkyl groups include methyl, ethyl, propyl, isopropyl, /7-butyl, f-butyl, isobutyl, pentyl, hexyl, isohexyl, heptyl, 4,4-dimethylpentyl, octyl and the like. Substituted alkyl groups include, but are not limited to, alkyl groups substituted by one or more of the following groups: halogen, hydroxy, alkanoyl, alkoxy, alkanoyloxy, thiol, alkylthio, alkylthiono, sulfonyl, sulfamoyl, carbamoyl, cyano, carboxy, acyl, aryl, alkenyl, alkynyl, aralkoxy, guanidino, optionally substituted amino, heterocyclyl including imidazolyl, furyl, thienyl, thiazolyl, pyrrolidyl, pyridyl, pyrimidyl and the like.
The term "lower alkyl" refers to those alkyl groups as described above having 1-7, preferably 2-4 carbon atoms.
The term "halogen" or "halo" refers to fluorine, chlorine, bromine and iodine.
The term "alkenyl" refers to any of the above alkyl groups having at least two carbon atoms and further containing a carbon to carbon double bond at the point of attachment. Groups having 2-4 carbon atoms are preferred. The term "alkynyl" refers to any of the above alkyl groups having at least two carbon atoms and further containing a carbon to carbon triple bond at the point of attachment. Groups having 2-4 carbon atoms are preferred.
The term "alkylene" refers to a straight-chain bridge of 3-6 carbon atoms connected by single bonds, e.g., -(CH2)X-, wherein x is 3-6, which may be interrupted with one or more heteroatoms selected from O, -O-C(O)-, S, S(O), S(O)2 or NR, wherein R may be hydrogen, alkyl, cycloalkyl, aryl, heterocyclyl, aralkyl, heteroaralkyl, acyl, carbamoyl, sulfonyl, alkoxycarbonyl, aryloxycarbonyl or aralkoxycarbonyl and the like; and the alkylene may further be substituted with one or more substituents selected from optionally substituted alkyl, cycloalkyl, aryl, heterocyclyl, oxo, halogen, hydroxy, carboxy, alkoxy, alkoxycarbonyl and the like.
The term "cycloalkyl" refers to optionally substituted monocyclic, bicyclic or tricyclic hydrocarbon groups of 3-12 carbon atoms, each of which may contain one or more carbon to carbon double bonds, or the cycloalkyl may be substituted by one or more substituents, such as alkyl, halo, oxo, hydroxy, alkoxy, alkanoyl, acylamino, carbamoyl, alkylamino, dialkylamino, thiol, alkylthio, cyano, carboxy, alkoxycarbonyl, sulfonyl, sulfonamido, sulfamoyl, heterocyclyl and the like.
Exemplary monocyclic hydrocarbon groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl and cyclohexenyl and the like.
Exemplary bicyclic hydrocarbon groups include bornyl, indyl, hexahydroindyl, tetrahydronaphthyl, decahydronaphthyl, bicyclo[2.1.1]hexyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.1 ]heptenyl, 6,6-dimethylbicyclo[3.1.1 ]heptyl, 2,6,6-trimethylbicyclo[3.1.1 ]heptyl, bicyclo[2.2.2]octyl and the like.
Exemplary tricyclic hydrocarbon groups include adamantyl and the like.
The term "alkoxy" refers to alkyl-O-.
The term "alkanoyl" refers to alkyl-C(O)-.
The term "alkanoyloxy" refers to alkyl-C(O)-O-.
The terms "alkylamino" and "dialkylamino" refer to alkyl-NH- and (alkyl)2N-, respectively. - A -
The term "alkanoylamino" refers to alkyl-C(O)-NH-.
The term "alkylthio" refers to alkyl-S-.
The term "trialkylsilyl" refers to (alkyl)3Si-.
The term "trialkylsilyloxy" refers to (alkyl)3Si0-.
The term "alkylthiono" refers to alkyl-S(O)-.
The term "alkylsulfonyl" refers to alkyl-S(O)2-.
The term "alkoxycarbonyl" refers to alkyl-O-C(O)-.
The term "alkoxycarbonyloxy" refers to alkyl-O-C(O)O-.
The term "carbamoyl" refers to H2NC(O)-, alkyl-NHC(O)-, (alkyl)2NC(O)-, aryl-NHC(O)-, alkyl(aryl)-NC(O)-, heteroaryl-N HC(O)-, alkyl(heteroaryl)-NC(O)-, aralkyl-NHC(O)-, alkyl(aralkyl)-NC(O)- and the like.
The term "sulfamoyl" refers to H2NS(O)2-, alkyl-NHS(O)2-, (alkyl)2NS(O)2-, aryl-NHS(O)2-, alkyl(aryl)-NS(O)2-, (aryl)2NS(O)2-, heteroaryl-NHS(O)2-, aralkyl-NHS(O)2-, heteroaralkyl- NHS(O)2- and the like.
The term "sulfonamido" refers to alkyl-S(O)2-NH-, ary 1-S(O)2-N H-, aralkyl-S(O)2-NH-, heteroaryl-S(O)2-NH-, heteroaralkyl-S(O)2-NH-, alkyl-S(O)2-N(alkyl)-, aryl-S(O)2-N(alkyl)-, aralkyl-S(O)2-N(alkyl)-, heteroaryl-S(O)2-N(alkyl)-, heteroaralkyl-S(O)2-N(alkyl)- and the like.
The term "sulfonyl" refers to alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aralkylsulfonyl, heteroaralkylsulfonyl and the like.
The term "optionally substituted amino" refers to an amino group which may optionally be substituted by substituents such as optionally substituted alkyl, acyl, sulfonyl, alkoxycarbonyl, cycloalkoxycarbonyl, aryloxycarbonyl, heteroaryloxycarbonyl, aralkoxycarbonyl, heteroaralkoxycarbonyl, carbamoyl and the like.
The term "aryl" refers to monocyclic or bicyclic aromatic hydrocarbon groups having 6-12 carbon atoms in the ring portion, such as phenyl, biphenyl, naphthyl and tetrahydronaphthyl, each of which may optionally be substituted by 1-4 substituents, such as optionally substituted alkyl, trifluoromethyl, cycloalkyl, halo, hydroxy, alkoxy, acyl, alkanoyloxy, aryloxy, optionally substituted amino, thiol, alkylthio, arylthio, nitro, cyano, carboxy, alkoxycarbonyl, carbamoyl, alkylthiono, sulfonyl, sulfonamido, heterocyclyl and the like.
The term "monocyclic aryl" refers to optionally substituted phenyl as described under aryl.
The term "aralkyl" refers to an aryl group bonded directly through an alkyl group, such as benzyl.
The term "aralkanoyl" refers to aralkyl-C(O)-.
The term "aralkylthio" refers to aralkyl-S-.
The term "aralkoxy" refers to an aryl group bonded directly through an alkoxy group.
The term "arylsulfonyl" refers to aryl-S(O)2-.
The term "arylthio" refers to aryl-S-.
The term "aroyl" refers to aryl-C(O)-.
The term "aroyloxy" refers to aryl-C(O)-O-.
The term "aroylamino" refers to aryl-C(O)-NH-.
The term "aryloxycarbonyl" refers to aryl-O-C(O)-.
The term "heterocyclyl" or "heterocyclo" refers to an optionally substituted, fully saturated or unsaturated, aromatic or nonaromatic cyclic group, e.g., which is a 4- to 7-membered monocyclic, 7- to 12-membered bicyclic or 10- to 15-membered tricyclic ring system, which has at least one heteroatom in at least one carbon atom-containing ring. Each ring of the heterocyclic group containing a heteroatom may have 1 , 2 or 3 heteroatoms selected from nitrogen atoms, oxygen atoms and sulfur atoms, where the nitrogen and sulfur heteroatoms may also optionally be oxidized. The heterocyclic group may be attached at a heteroatom or a carbon atom.
Exemplary monocyclic heterocyclic groups include pyrrolidinyl, pyrrolyl, pyrazolyl, oxetanyl, pyrazolinyl, imidazolyl, imidazolinyl, imidazolidinyl, triazolyl, oxazolyl, oxazolidinyl, isoxazolinyl, isoxazolyl, thiazolyl, thiadiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, furyl, tetrahydrofuryl, thienyl, oxadiazolyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolodinyl, 2-oxoazepinyl, azepinyl, 4-piperidonyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, tetrahydropyranyl, morpholinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiamorpholinyl sulfone, 1 ,3-dioxolane and tetrahydro-1 ,1- dioxothienyl, 1 ,1 ,4-trioxo-1 ,2,5-thiadiazolidin-2-yl and the like.
Exemplary bicyclic heterocyclic groups include indolyl, dihydroidolyl, benzothiazolyl, 4,5,6,7- tetrahydro-benzothiazolyl, benzoxazinyl, benzoxazolyl, benzothienyl, benzothiazinyl, thiazolo[5,4-b]pyridinyl, thiazolo[5,4-d]pyrimidinyl, oxazolo[5,4-b]pyridinyl, 6,7-dihydro-4H- thiopyrano[4,3-d]thiazolyl, 6,7-dihydro-4H-pyrano[4,3-d]thiazolyl, 4,5,6,7-tetrahydro- thiazolo[5,4-c]pyridinyl, 4,5,6,7-tetrahydro-pyrazolo[1 ,5-a]pyridinyl, 5,6,7,8-tetrahydro- triazolo[1 ,5-a]pyridinyl, quinuclidinyl, quinolinyl, tetrahydroquinolinyl, decahydroquinolinyl, isoquinolinyl, tetrahydroisoquinolinyl, decahydroisoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuryl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridinyl (such as furo[2,3-c]pyridinyl, furo[3,2-b]-pyridinyl] or furo[2,3-b]pyridinyl), dihydroisoindolyl, 1 ,3-dioxo-1 ,3-dihydroisoindol-2-yl, dihydroquinazolinyl (such as 3,4-dihydro-4-oxo-quinazolinyl), phthalazinyl and the like.
Exemplary tricyclic heterocyclic groups include carbazolyl, dibenzoazepinyl, dithienoazepinyl, benzindolyl, phenanthrolinyl, acridinyl, phenanthridinyl, phenoxazinyl, phenothiazinyl, xanthenyl, carbolinyl and the like.
The term "heterocyclyl" includes substituted heterocyclic groups. Substituted heterocyclic groups refer to heterocyclic groups substituted with 1 , 2 or 3 substituents selected from the group consisting of the following:
(a) optionally substituted alkyl;
(b) hydroxy! (or protected hydroxyl);
(c) halo;
(d) oxo, i.e., =0;
(e) optionally substituted amino;
(f) alkoxy;
(g) cycloalkyl;
(h) free or esterified carboxy; (i) heterocyclyl; G) alkylthio; (k) alkylthiono;
(I) nitro;
(m) cyano;
(n) sulfamoyl;
(o) alkanoyloxy;
(p) aroyloxy;
(q) arylthio;
(r) aryloxy;
(s) sulfamoyl;
(t) sulfonyl;
(u) carbamoyl;
(v) aralkyl; and
(w) aryl optionally substituted with alkyl, cycloalkyl, alkoxy, hydroxyl, amino, acylamino, alkylamino, dialkylamino or halo.
The term "heterocyclooxy" denotes a heterocyclic group bonded through an oxygen bridge.
The term "heteroaryl" refers to an aromatic heterocycle, e.g., monocyclic or bicyclic aryl, such as pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, furyl, thienyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolyl, benzothiazolyl, benzoxazolyl, benzothienyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzofuryl and the like, optionally substituted by, e.g., lower alkyl, lower alkoxy or halo.
The term "heteroarylsulfonyl" refers to heteroaryl-S(O)2-.
The term "heteroaroyl" refers to heteroaryl-C(O)-.
The term "heteroaryloxycarbonyl" refers to heteroaryl-O-C(O)-.
The term "heteroaroylamino" refers to heteroaryl-C(O)NH-.
The term "heteroaralkyl" refers to a heteroaryl group bonded through an alkyl group.
The term "heteroaralkanoyl" refers to heteroaralkyl-C(O)-.
The term "heteroaralkanoylamino" refers to heteroaralkyl-C<O)NH-. The term "acyl" refers to alkanoyl, aroyl, heteroaroyl, aralkanoyl, heteroaralkanoyl and the like.
The term "acylamino" refers to alkanoylamino, aroylamino, heteroaroylamino, aralkanoylamino, heteroaralkanoylamino and the like.
The term "esterified carboxy" refers to optionally substituted alkoxycarbonyl, cycloalkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, heterocyclooxycarbonyl and the like.
Pharmaceutically acceptable salts of the compounds of the present invention refer to salts formed with acids, namely acid addition salts, such as of mineral acids, organic carboxylic acids and organic sulfonic acids, e.g., hydrochloric acid, maleic acid and methanesulfonic acid, respectively.
Similarly, pharmaceutically acceptable salts of the compounds of the invention refer to salts formed with bases, namely cationic salts, such as alkali and alkaline earth metal salts, e.g., sodium, lithium, potassium, calcium and magnesium, as well as ammonium salts, e.g., ammonium, trimethylammonium, diethylammonium and tris(hydroxymethyl)-methyl- ammonium salts and salts with amino acids provided an acidic group constitutes part of the structure.
As described herein above, the present invention provides certain sulfonamide derivatives of formula (I), pharmaceutical compositions containing them, methods for preparing said compounds, and methods of treating glucokinase mediated conditions by administration of a therapeutically effective amount of a compound of the present invention, or a pharmaceutical composition thereof.
Preferred are the compounds of formula (I) wherein R3 is cyclopentyl; R4 is hydrogen; R6 is free or esterified carboxy; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
Further preferred are the compounds of formula (I), designated as the A group, having the formula
Figure imgf000010_0001
wherein
R9 is hydrogen, optionally substituted lower alkyl, lower alkenyl, cycloalkyl, aryl or aralkyl;
Q combined together with the carbon and nitrogen atoms to which it is attached form a 5- to 6-membered monocyclic heteroaromatic ring which is selected from the group consisting of
Figure imgf000010_0002
or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
Preferred are the compounds in the A group wherein n is an integer from 1 to 3; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
/
More preferred are the compounds in the A group wherein n is an integer from 1 to 3; R9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
More preferred are also the compounds in the A group wherein n is an integer from 1 to 3; R1 is hydrogen, halogen, cyano, trifluoromethyl, alkoxy, alkylthio or carboxy; R2 is absent; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
Most preferred are the compounds in the A group, designated as the B group, wherein n is an integer from 1 to 3;
R1 is hydrogen, halogen, cyanb, trifluoromethyl, alkoxy, alkylthio or carboxy; R2 is absent;
Q combined together with the carbon and nitrogen atoms to which it is attached form a 5- to 6-membered monocyclic heteroaromatic ring which is selected from the group consisting of
Figure imgf000011_0001
or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
Preferred are the compounds in the B group wherein
R5 is (C3-7)cycloalkyl or (C1-4)alkyl substituted by (C1-6)alkoxy, (C3-7)cycloalkyl, (C6-1o)aryl or (C5-6)heterocyclyl; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
More preferred are the compounds in the B group wherein
R5 is (C3-7)cycloalkyl or (C1-4)alkyl substituted by (C1-6)alkoxy, (C3.7)cycloalkyl, (C6-i0)aryl or (C5-6)heterocyclyl;
R9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
Most preferred are the compounds in the B group wherein R5 is (C3.7)cycloalkyl or (C^alkyl substituted by (Ci-6)alkoxy, (C3-7)cycloalkyl, (C6-io)aryl or (C5.6)heterocyclyl in which (Ci-4)alkyl is methyl or ethyl, (C1-6)alkoxy is methoxy or ethoxy; and (C6-io)aryl is optionally substituted phenyl;
R9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
Further preferred are also the compounds of formula (I), designated as the C group, having the formula
Figure imgf000012_0001
wherein
R9 is hydrogen, optionally substituted lower alkyl, lower alkenyl, cycloalkyl, aryl or aralkyl;
Q combined together with the carbon and nitrogen atoms to which it is attached form a 9- to 10-membered bicyclic heterocycle which is selected from the group consisting of
Figure imgf000012_0002
Figure imgf000013_0001
or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
Preferred are the compounds in the C group wherein n is an integer from 1 to 3; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
More preferred are the compounds in the C group wherein n is an integer from 1 to 3; R9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
More preferred are also the compounds in the C group wherein n is an integer from 1 to 3;
R1 is hydrogen, halogen, cyano, trifluoromethyl, alkoxy, alkylthio or carboxy; R2 is absent; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
Most preferred are the compounds in the C group, designated as the D group, wherein n is an integer from 1 to 3;
R1 is hydrogen, halogen, cyano, trifluoromethyl, alkoxy, alkylthio or carboxy; R2 is absent;
Q combined together with the carbon and nitrogen atoms to which it is attached form a 9- to 10-membered bicyclic heterocycle which is selected from the group consisting of
Figure imgf000014_0001
or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
Preferred are the compounds in the D group wherein
R5 is (C3.7)cycloalkyl or (C1-4)alkyl substituted by (C1-6)alkoxy, (C3-7)cycloalkyl, (C6-10)aryl or (C5.6)heterocyclyl; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
More preferred are the compounds in the D group wherein
R5 is (C3.7)cycloalkyl or (C1-4)alkyl substituted by (C1-6)alkoxy, (C3-7)cycloalkyl, (C6-io)aryl or (C5.6)heterocyclyl;
R9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
Most preferred are the compounds in the D group wherein
R5 is (C3-7)cycloalkyl or (C-|.4)alkyl substituted by (Ci-6)alkoxy, (C3-7)cycloalkyl, (C6-io)aryl or (C5-6)heterocyclyl in which (C1-4)alkyl is methyl or ethyl, (C-ι.6)alkoxy is methoxy or ethoxy; and (C6-io)aryI is optionally substituted phenyl;
R9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
The compounds of the invention depending on the nature of the substituents possess one or more asymmetric centers. The resulting diastereoisomers, optical isomers, i.e., enantiomers, and geometric isomers, and mixtures thereof, are encompassed by the instant invention. Preferred are the compounds of the present invention wherein the substituent at the carbon atom adjacent to the amide group attains the R-configuration. Particular embodiments of the invention are:
3-(Benzyl-{4-[(R)-2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-6]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-amino)-propionic acid;
3-[{4-[2-Cyclopentyl-1-([1 ,3,4]thiadiazol-2-ylcarbamoyl)-ethyl]-benzene-sulfonyl}-(2-methoxy- ethyl)-amino]-propionic acid;
3-(Benzofuran-2-ylmethyl-{4-[(R)-2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-ό]pyridin-2- ylcarbamoyO-ethyO-benzenesulfonylJ-aminoJ-propionic acid;
3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-/?]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-pyridin-2-ylmethyl-amino)-propionic acid;
3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-ib]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-oxazol-2-ylmethyl-amino)-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-ό]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(tetrahydro-furan-2-ylmethyl)-amino]-propionic acid;
3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-thiophen-2-ylmethyl-amino)-propionic acid;
4-[((2-Carboxy-ethyl)-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-ό]pyridin-2-ylcarbamoyl)- ethyll-benzenesulfonylj-aminoj-methylj-benzoic acid;
3-(Cyclohexylmethyl-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-jb]pyridin-2-ylcarbamoyl)- ethyl]-benzenesulfonyl}-amino)-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-ό]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(5-methyl-furan-2-ylmethyl)-amino]-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-ib]pyridin-2-ylcarbamoyl)-etriyl]- benzenesulfonyl}-(2,5-dimethyl-furan-3-ylmethyl)-amino]-propionic acid;
[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-ό]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(5-methyl-furan-2-ylmethyl)-amino]-acetic acid;
3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-/)]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-isopropyl-amino)-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-ϋ]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid; 3-[{4-[2-Cyclopentyl-1-(5-methoxy-1 ,2-dihydro-thiazolo[5,4-jb]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(1 ,5-dimethyl-1 /-/-pyrazol-S-ylmethyO-aminoJ-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-ό]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(5-methyl-isoxazol-3-ylmethyl)-amino]-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-/)]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(tetrahydro-pyran-4-ylmethyl)-amino]-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-/?]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(4-fluoro-benzyl)-amino]-propionic acid;
3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-/)]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-cyclopropylmethyl-amino)-propionic acid;
({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-ό]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyty-methyl-aminoj-acetic acid;
3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-ib]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-methyl-amino)-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-jb]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl^Ctetrahydro-pyran^-ylJ-aminoJ-propionic acid;
3-(Cyclohexyl-{4-[(S)-2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-jb]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonylj-aminoj-propionic acid;
3-[{4-[(S)-2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-ό]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(4-methoxy-phenyl)-amino]-propionic acid;
3-({4-[(S)-2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-/b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-methyl-amino)-propionic acid;
3-[{4-[(S)-2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-ib]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-isopropoxy-ethyl)-amino]-propionic acid;
3-({4-[(S)-2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-ύ]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonylHhiazol^-ylmethyl-aminoJ-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-fluoro-pyridin-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2-methoxy- ethyl)-amino]-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(pyrimidin-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2-methoxy- ethyl)-amino]-propionic acid; 3-[{4-[(R)-2-Cyclopentyl-1-(4-methyl-thia2ol-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-2- methoxy-ethyO-aminoJ-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1 -(1 -methyl- 1 H-pyrazol-3-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid; and
3-[{4-[(R)-2-Cyclopentyl-1-(pyrimidin-4-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2-methoxy- ethyl)-amino]-propionic acid; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
Compounds of formula (I) may be prepared using methods well known in the art, e.g., according to Method A or Method B as outlined herein below.
Method A:
Compounds of formula (I) may be obtained by coupling an amine of the formula
Figure imgf000017_0001
or acid addition salts thereof, wherein FV and R2' represents R1 and R2, respectively, as defined herein above, or R1' and R2 1 are groups convertible to R1 and R2, respectively, with an activated derivative of a carboxylic acid of the formula
Figure imgf000017_0002
wherein R3, R4 and n have meanings as defined herein, and R5' and R6' represents R5 and R6, respectively, as defined herein above, or R5' and R6' are groups convertible to R5 and R6, respectively, to afford a compound of the formula
Figure imgf000018_0001
wherein R1', R2', R3, R4, n, R5' and R6' have meanings as defined for formulae (II) and (III).
In the coupling reaction cited herein above, an activated derivative of a carboxylic acid, e.g., those corresponding to carboxylic acids of formula (III), include acid chlorides, bromides and fluorides, mixed anhydrides, lower alkyl esters and activated esters thereof, and adducts formed with coupling agents, such as 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDCI), 1-hydroxy benzotriazole (HOBt), 0-(1 ,2-dihydro-2-oxo-1-pyridyl)- N, N, N ', N -tetramethyluronium tetrafluoroborate, benzotriazole-1 -yl-oxy-tris-pyrrolidino- phosphonium hexafluorophosphate (PyBOP) and the like. Mixed anhydrides are preferably such from pivalic acid, or lower alkyl hemiesters of carbonic acids, such as ethyl or isobutyl analogs. Activated esters include, for example, succinimido, phthalimido or 4-nitrophenyl esters. The reaction of an activated derivative of a carboxylic acid, e.g., those corresponding to carboxylic acids of formula (III), with an amine, e.g., those of formula (II), may be carried out in the presence of a base, such as pyridine, triethylamine (TEA), diisopropylethylamine (DIEA) or N-methylmorpholine (NMM) in an inert organic solvent, such as dichloromethane (DCM), N,N-dimethylformamide (DMF) or tetrahydrofuran (THF), or a mixture of solvents thereof. Carboxylic acids of formula (III) may be converted to their activated derivatives using methods described herein or according to methods generally known in the art, e.g., a carboxylic acid of formula (III) may be treated with a chlorinating agent, such as thionyl chloride or oxalyl chloride, to afford a corresponding acid chloride thereof, or by the treatment of a coupling agent such as EDCI or HOBt, or a mixture of coupling agents thereof.
Amines of formula (II) and carboxylic acids of formula (III) are known, or if they are novel they may be prepared using methods well known in the art or as described herein in the illustrative Examples, or modifications thereof. For example, compounds of formula (III) may be prepared by treating an ester of the formula
Figure imgf000019_0001
wherein R4 has a meaning as defined herein above, and R is lower alkyl, preferably, methyl or ethyl, with chlorosulfonic acid to afford a compound of the formula
Figure imgf000019_0002
wherein R4 and R have meanings as defined herein above, optionally in the presence of an intrinsic organic solvent. Preferably, the reaction is carried out without an intrinsic organic solvent.
A compound of formula (V) may then be treated with an amine of the formula
R6!-(CH2)n-NH-R5' (Vl), or an acid addition salt thereof, wherein n, R5' and R6' have meanings as defined herein above, in the presence of a base, such as pyridine, TEA, DIEA or NMM, in an inert organic solvent, such as DCM, DMF or THF, or a mixture of solvents thereof, to afford a compound of the formula
Figure imgf000019_0003
wherein R4, n, R5', R6' and R have meanings as defined herein above. Preferably, the reaction is conducted at a temperature ranging from about -40C to room temperature (RT), more preferably, the reaction temperature is about O0C. Amines of formula (Vl) are known, or if they are novel they may be prepared using methods well known in the art or as described herein in the illustrative Examples.
A resulting compound of formula (VII) may then be treated with a base, such as sodium hydride, lithium diisopropylamide (LDA) or lithium bis(trimethylsilyl)amide (LHMDS), preferably LDA, followed by addition of an alkylating agent of the formula R3-(CH2)-Lg (VIII) wherein R3 has a meaning as defined herein above, and Lg represents a leaving group, such as chloride, bromide, iodide, mesylate, tosylate or triflate, preferably iodide ot triflate, to afford a compound of the formula
Figure imgf000020_0001
wherein R3, R4, n, R5', R6' and R have meanings as defined herein above. The alkylation step is preferably conducted in a polar organic solvent, such as THF, DMF, N-methylpyrrolidone (NMP), 1 ,3-dimethyl-3,4,5,6-tetrahydro-2('//-/)-pyridone (DMPU) or 1 ,3- dimethyl-3,4,5,6-tetrahydro-2(7/-/)-pyrimidinone (DMTP), or in a mixture of solvents thereof.
A resulting compound of formula (IX) may then be hydrolyzed, e.g., in the presence of an aqueous base such as sodium, lithium or potassium hydroxide and an organic solvent such as THF or lower alcohol, preferably, methanol or ethanol, to afford a carboxylic acid of formula (III) wherein R3, R4, n, R5' and R6' have meanings as defined herein above.
A carboxylic acid of formula (III) may then be coupled with an amine of formula (II), or an acid addition salt thereof, under reaction conditions as described herein above to afford a compound of formula (I1) wherein R1', R2', R3, R4, n, R5' and R6' have meanings as defined herein above, e.g., via conversion of the acid to the corresponding acid chloride or in the presence of a coupling agent such as EDCI, HOBt or PyBOP, or a mixture of coupling agents thereof.
Alternatively, compounds of formula (I) may be prepared as outlined herein below.
Method B:
Compounds of formula (I) may be obtained by reacting a compound of the formula
Figure imgf000021_0001
wherein R3 and R4 have meanings as defined herein above, and R1' and R2' represents Ri and R2, respectively, as defined herein above, or R1' and R2' are a groups convertible to Ri and R2, respectively, with an amine of the formula
R6'-(CH2)n-NH-R5' (Vl), or an acid addition salt thereof, wherein n, R5' and R6' have meanings as defined herein above, in the presence of a base, such as pyridine, TEA, DIEA or NMM, in an inert organic solvent, such as DCM, DMF or THF, or a mixture of solvents thereof, to afford a compound of the formula
Figure imgf000021_0002
wherein R1', R2', R3, R4, n, R5' and R6' have meanings as defined herein above.
Compounds of formula (X) may be prepared, e.g., by treating a compound of the formula
Figure imgf000021_0003
wherein R4 and R have meanings as defined herein above, with a base, such as sodium hydride, LDA or LHMDS, preferably LDA, followed by addition of an alkylating agent of the formula
R3-(CH2)-Lg (VIII) wherein R3 has a meaning as defined herein above, and Lg represents a leaving group, such as chloride, bromide, iodide, mesylate, tosylate or triflate, preferably iodide or triflate, to afford a compound of the formula
Figure imgf000022_0001
wherein R3, R4 and R have meanings as defined herein above. The alkylation step is preferably conducted in a polar organic solvent, such as THF, DMF, NMP, DMPU or DMTP, or in a mixture of solvents thereof.
A resulting compound of formula (XII) may then be hydrolyzed, e.g., in the presence of an aqueous base, such as sodium, lithium or potassium hydroxide and an organic solvent such as THF or lower alcohol, preferably, methanol or ethanol, to afford a carboxylic acid of the formula
Figure imgf000022_0002
wherein R3 and R4 have meanings as defined herein above.
A carboxylic acid of formula (XIII) may then be coupled with an amine of formula (II) under reaction conditions as described herein above to afford a compound of the formula
Figure imgf000022_0003
wherein R1', R2', R3 and R4 have meanings as defined herein above, e.g., via conversion of the carboxylic acid to the corresponding acid chloride, or in the presence of a coupling agent, such as EDCI, HOBt or PyBOP, or a mixture of coupling agents thereof. A resulting compound of formula (XIV) may then be converted to a sulfonyl chloride derivative of formula (X) wherein R1', R2', R3 and R4 have meanings as defined herein above, by reduction of the nitro group to the amino group, e.g., using iron powder in the presence of a mixture of acetic acid and a lower alcohol, such as ethanol, followed by diazotization reaction and subsequent treatment with, e.g., sulfur dioxide in the presence of copper(ll) chloride and acetic acid.
A resulting sulfonyl chloride derivative of formula (X) may then be treated with an amine of formula (Vl), or an acid addition salt thereof, wherein n, R5' and R6' have meanings as defined herein above, under reaction conditions described herein above to afford a compound of formula (I1) wherein R1', R2', R3, R4, n, R5' and R6' have meanings as defined herein above.
The processes described herein above may be conducted under inert atmosphere, preferably under nitrogen atmosphere.
In starting compounds and intermediates which are converted to the compounds of the present invention in a manner described herein, functional groups present, such as amino, thiol, carboxyl and hydroxyl groups, are optionally protected by conventional protecting groups that are common in preparative organic chemistry. Protected amino, thiol, carboxyl and hydroxyl groups are those that can be converted under mild conditions into free amino thiol, carboxyl and hydroxyl groups without the molecular framework being destroyed or other undesired side reactions taking place.
The purpose of introducing protecting groups is to protect the functional groups from undesired reactions with reaction components under the conditions used for carrying out a desired chemical transformation. The need and choice of protecting groups for a particular reaction is known to those skilled in the art and depends on the nature of the functional group to be protected (hydroxyl group, amino group, etc.), the structure and stability of the molecule of which the substituent is a part and the reaction conditions.
Well-known protecting groups that meet these conditions and their introduction and removal are described, e.g., in McOmie, "Protective Groups in Organic Chemistry", Plenum Press, London, NY (1973); and Greene and Wuts, "Protective Groups in Organic Synthesis", John Wiley and Sons, Inc., NY (1999). The above-mentioned reactions are carried out according to standard methods, in the presence or absence of diluent, preferably, such as are inert to the reagents and are solvents thereof, of catalysts, condensing or said other agents, respectively and/or inert atmospheres, at low temperatures, RT or elevated temperatures, preferably at or near the boiling point of the solvents used, and at atmospheric or super-atmospheric pressure. The preferred solvents, catalysts and reaction conditions are set forth in the appended illustrative Examples.
The invention further includes any variant of the present processes, in which an intermediate product obtainable at any stage thereof is used as starting material and the remaining steps are carried out, or in which the starting materials are formed in situ under the reaction conditions, or in which the reaction components are used in the form of their salts or optically pure antipodes.
Compounds of the invention and intermediates can also be converted into each other according to methods generally known perse.
The invention also relates to any novel starting materials, intermediates and processes for their manufacture.
Depending on the choice of starting materials and methods, the new compounds may be in the form of one of the possible isomers or mixtures thereof, for example, as substantially pure geometric (cis or trans) isomers, diastereomers, optical isomers (enantiomers, antipodes), racemates or mixtures thereof. The aforesaid possible isomers or mixtures thereof are within the purview of this invention.
Any resulting mixtures of isomers can be separated on the basis of the physicochemical differences of the constituents, into the pure geometric or optical isomers, diastereomers, racemates, for example, by chromatography and/or fractional crystallization.
Any resulting racemates of final products or intermediate, e.g., acids of formulae (III) and (XIII), can be resolved into the optically pure isomers by known methods, e.g., by separation of the diastereomeric salts thereof, obtainable with an optically active acid or base and liberating the optically active acidic or basic compound, respectively, e.g., acids of formulae (III) and (XIII) can be resolved using optically active 1-phenylethylamine. Similarly, the compounds of the instant invention having a basic moiety may be resolved into their optical isomers, e.g., by fractional crystallization of a salt formed with an optically active acid, e.g., tartaric acid, dibenzoyl tartaric acid, diacetyl tartaric acid, di-0,0'-p-toluoyl tartaric acid, mandelic acid, malic acid or camphor-10-sulfonic acid. Racemic products can also be resolved by chiral chromatography, e.g., high pressure liquid chromatography (HPLC) using a chiral adsorbent.
Alternatively, optically pure isomers of compounds of the present invention may be obtained by employing chiral reagents. For example, an optical isomer, preferably the R isomer, of compounds of formula (X) may be prepared employing chiral auxiliaries, e.g., the Evans auxiliary, as illustrated herein in the Examples.
Finally, compounds of the invention are either obtained in the free form, or in a salt form thereof, preferably, in a pharmaceutically acceptable salt form thereof, or as a prodrug derivative thereof.
Compounds of the instant invention which contain acidic groups may be converted into salts with pharmaceutically acceptable bases. Such salts include alkali metal salts, like sodium, lithium and potassium salts; alkaline earth metal salts, like calcium and magnesium salts; ammonium salts with organic bases, e.g., trimethylamine salts, diethylamine salts, tris(hydroxymethyl)methylamine salts, dicyclohexylamine salts and N-methyl-D-glucamine salts; salts with amino acids like arginine, lysine and the like. Salts may be formed using conventional methods, advantageously in the presence of an ethereal or alcoholic solvent, such as a lower alkanol. From the solutions of the latter, the salts may be precipitated with ethers, e.g., diethyl ether. Resulting salts may be converted into the free compounds by treatment with acids. These or other salts can also be used for purification of the compounds obtained.
Compounds of the invention, in general, may be converted into acid addition salts, especially pharmaceutically acceptable salts. These are formed, e.g., with inorganic acids, such as mineral acids, e.g., sulfuric acid, phosphoric or hydrohalic acid, or with organic carboxylic acids, such as (Ci-4)alkanecarboxylic acids which, e.g., are unsubstituted or substituted by halogen, e.g., acetic acid, such as saturated or unsaturated dicarboxylic acids, e.g., oxalic, succinic, maleic or fumaric acid, such as hydroxycarboxylic acids, e.g., glycolic, lactic, malic, tartaric or citric acid, such as amino acids, e.g., aspartic or glutamic acid, or with organic sulfonic acids, such as (Cr4)alkylsulfonic acids, e.g., methanesulfonic acid; or arylsulfonic acids which are unsubstituted or substituted (for example by halogen). Preferred are salts formed with hydrochloric acid, maleic acid and methanesulfonic acid. Prodrug derivatives of any compound of the invention are derivatives of said compounds which following administration release the parent compound in vivo via some chemical or physiological process, e.g., a prodrug on being brought to the physiological pH or through enzyme action is converted to the parent compound. Exemplary prodrug derivatives are, e.g., esters of free carboxylic acids and S-acyl and O-acyl derivatives of thiols, alcohols or phenols, wherein acyl has a meaning as defined herein. Preferred are pharmaceutically acceptable ester derivatives convertible by solvolysis under physiological conditions to the parent carboxylic acid. Such ester derivatives include, but are not limited to, lower alkyl esters, cycloalkyl esters, lower alkenyl esters, benzyl esters, mono- or di-substituted lower alkyl esters, such as the ω-(amino, mono- or di-lower alkylamino, carboxy, lower alkoxycarbonyl)-lower alkyl esters, the α-(lower alkanoyloxy, lower alkoxycarbonyl or di-lower alkylaminocarbonyl)-lower alkyl esters, such as the pivaloyloxymethyl ester and others conventionally used in the art, e.g., a compound of formula (IA) wherein R9 is optionally substituted lower alkyl as described herein above, lower alkenyl, cycloalkyl, aryl or aralkyl, is easily convertible to a compound of formula (IA) wherein R9 is hydrogen.
In view of the close relationship between the free compounds, the prodrug derivatives and the compounds in the form of their salts, whenever a compound is referred to in this context, a prodrug derivative and a corresponding salt is also intended, provided such is possible or appropriate under the circumstances.
The compounds, including their salts, can also be obtained in the form of their hydrates, or include other solvents used for their crystallization.
As described herein above, the compounds of the present invention may be employed for the treatment of conditions mediated by glucokinase activity. Such compounds may thus be employed therapeutically for the treatment of impaired glucose tolerance, type 2 diabetes and obesity.
The present invention further provides pharmaceutical compositions comprising a therapeutically effective amount of a pharmacologically active compound of the instant invention, alone or in combination with one or more pharmaceutically acceptable carriers.
The pharmaceutical compositions according to the invention are those suitable for enteral, such as oral or rectal; transdermal and parenteral administration to mammals, including man, for the treatment of conditions mediated by glucokinase activity. Such conditions include impaired glucose tolerance, type 2 diabetes and obesity.
Thus, the pharmacologically active compounds of the invention may be employed in the manufacture of pharmaceutical compositions comprising an effective amount thereof in conjunction or admixture with excipients or carriers suitable for either enteral or parenteral application. Preferred are tablets and gelatin capsules comprising the active ingredient together with: a) diluents, e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) lubricants, e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also c) binders, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and or polyvinylpyrrolidone; if desired d) disintegrants, e.g., starches, agar, alginic acid or its sodium salt, or effervescent mixtures; and/or e) absorbants, colorants, flavors and sweeteners.
Injectable compositions are preferably aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions.
Said compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. In addition, they may also contain other therapeutically valuable substances. Said compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1-75%, preferably about 1-50%, of the active ingredient.
Suitable formulations for transdermal application include a therapeutically effective amount' of a compound of the invention with carrier. Advantageous carriers include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host. Characteristically, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound of the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin. Accordingly, the present invention provides pharmaceutical compositions as described above for the treatment of conditions mediated by glucokinase activity, preferably, impaired glucose tolerance, type 2 diabetes and obesity.
The pharmaceutical compositions may contain a therapeutically effective amount of a compound of the invention as defined above, either alone or in a combination with another therapeutic agent, e.g., each at an effective therapeutic dose as reported in the art. Such therapeutic agents include: a) anti-diabetic agents, such as insulin, insulin derivatives and mimetics; insulin secretagogues such as the sulfonylureas, e.g., Glipizide, glyburide and Amaryl; insulinotropic sulfonylurea receptor ligands such as meglitinides, e.g., nateglinide and repaglinide; thiazolidone derivatives such as glitazones, e.g., pioglitazone and rosiglitazone; protein tyrosine phosphatase-1 B (PTP-1 B) inhibitors such as PTP-112; GSK3 (glycogen synthase kinase-3) inhibitors such as SB-517955, SB-4195052, SB-216763, NN-57-05441 and NN-57- 05445; RXR ligands such as GW-0791 and AGN-194204; sodium-dependent glucose co- transporter inhibitors such as T-1095; glycogen phosphorylase A inhibitors such as BAY R3401 ; biguanides such as metformin; alpha-glucosidase inhibitors such as acarbose; GLP- 1 (glucagon like peptide-1), GLP-1 analogs such as Exendin-4 and GLP-1 mimetics; modulators of PPARs (peroxisome proliferator-activated receptors), e.g., non-glitazone type PPARγ agonists such as N-(2-benzoylphenyl)-L-tyrosine analogues, e.g. GI-262570, and JTT501 ; DPPIV (dipeptidyl peptidase IV) inhibitors such as LAF237, MK-0431 , saxagliptin and GSK23A; SCD-1 (stearoyl-CoA desaturase-1) inhibitors; DGAT1 and DGAT2 (diacylglycerol acyltransferase 1 and 2) inhibitors; ACC2 (acetyl CoA carboxylase 2) inhibitors; and breakers of AGE (advanced glycation end products); b) anti-dyslipidemic agents such as 3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase inhibitors, e.g., lovastatin, pitavastatin, simvastatin, pravastatin, cerivastatin, mevastatin, velostatin, fluvastatin, dalvastatin, atorvastatin, rosuvastatin and rivastatin; HDL increasing compounds such as cholesterol ester transfer protein (CETP) inhibitors, e.g., JTT705; Apo-A1 analogs and mimetics; squalene synthase inhibitors; FXR (farnesoid X receptor) and LXR (liver X receptor) ligands; cholestyramine; fibrates; nicotinic acid; and aspirin; c) anti-obesity agents such as phentermine, leptin, bromocriptine, dexamphetamine, amphetamine, fenfluramine, dexfenfluramine, sibutramine, orlistat, dexfenfluramine, mazindol, phentermine, phendimetrazine, diethylpropion, fluoxetine, bupropion, topiramate, diethylpropion, benzphetamine, phenylpropanolamine, ecopipam, ephedrine, and pseudoephedrine; cholesterol absorption modulators such as ZETIA® and KT6-971 ; and cannabinoid receptor antagonists such as rimonabant; and d) anti-hypertensive agents, e.g., loop diuretics such as ethacrynic acid, furosemide and torsemide; angiotensin converting enzyme (ACE) inhibitors such as benazepril, captopril, enalapril, fosinopril, lisinopril, moexipril, perinodopril, quinapril, ramipril and trandolapril; inhibitors of the Na-K-ATPase membrane pump such as digoxin; neutralendopeptidase (NEP) inhibitors; ACE/NEP inhibitors such as omapatrilat, sampatrilat and fasidotril; angiotensin Il antagonists such as candesartan, eprosartan, irbesartan, losartan, telmisartan and valsartan, in particular valsartan; renin inhibitors such as ditekiren, zankiren, terlakiren, aliskiren, RO 66-1132 and RO-66-1168; β-adrenergic receptor blockers such as acebutolol, atenolol, betaxolol, bisoprolol, metoprolol, nadolol, propranolol, sotalol and timolol; inotropic agents such as digoxin, dobutamine and milrinone; calcium channel blockers such as amlodipine, bepridil, diltiazem, felodipine, nicardipine, nimodipine, nifedipine, nisoldipine and verapamil; aldosterone receptor antagonists such as eplerenone; and aldosterone synthase inhibitors such as anastrazole and fadrazole.
Other specific anti-diabetic compounds are described by Patel Mona in Expert Opin Investig Drugs, 2003, 12(4), 623-633, in the figures 1 to 7, which are herein incorporated by reference. A compound of the present invention may be administered either simultaneously, before or after the other active ingredient, either separately by the same or different route of administration or together in the same pharmaceutical formulation.
The structure of the therapeutic agents identified by code numbers, generic or trade names may be taken from the actual edition of the standard compendium "The Merck Index" or from databases, e.g., Patents International (e.g. IMS World Publications). The corresponding content thereof is hereby incorporated by reference.
Accordingly, the present invention provides pharmaceutical compositions comprising a therapeutically effective amount of a compound of the invention in combination with a therapeutically effective amount of another therapeutic agent, preferably selected from antidiabetics, hypolipidemic agents, anti-obesity agents or anti-hypertensive agents, most preferably from antidiabetics or hypolipidemic agents as described above.
The present invention further relates to pharmaceutical compositions as described above for use as a medicament. The present invention further relates to use of pharmaceutical compositions or combinations as described above for the preparation of a medicament for the treatment of conditions mediated by glucokinase activity, preferably, impaired glucose tolerance, type 2 diabetes and obesity.
Thus, the present invention also relates to a compound of formula (I) for use as a medicament; to the use of a compound of formula (I) for the preparation of a pharmaceutical composition for the prevention and/or treatment of conditions mediated by glucokinase activity, and to a pharmaceutical composition for use in conditions mediated by glucokinase activity comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable diluent or carrier therefore.
The present invention further provides a method for the prevention and/or treatment of conditions mediated by glucokinase activity, which comprises administering a therapeutically effective amount of a compound of the present invention.
A unit dosage for a mammal of about 50-70 kg may contain between about 1 mg and 1000 mg, advantageously between about 5-500 mg of the active ingredient. The therapeutically effective dosage of active compound is dependent on the species of warm-blooded animal (mammal), the body weight, age and individual condition, on the form of administration, and on the compound involved.
In accordance with the foregoing the present invention also provides a therapeutic combination, e.g., a kit, kit of parts, e.g., for use in any method as defined herein, comprising a compound of formula (I), or a pharmaceuticallyacceptable salt thereof, to be used concomitantly or in sequence with at least one pharmaceutical composition comprising at least another therapeutic agent, preferably selected from anti-diabetic agents, hypolipidemic agents, anti-obesity agents and anti-hypertensive agents, or a pharmaceutically acceptable salt thereof. The kit may comprise instructions for its administration.
Similarly, the present invention provides a kit of parts comprising: (i) a pharmaceutical composition of the invention; and (ii) a pharmaceutical composition comprising a compound selected from an anti-diabetic, a hypolipidemic agent, an anti-obesity agent and an antihypertensive agent, or a pharmaceutically acceptable salt thereof, in the form of two separate units of the components (i) to (ii). Likewise, the present invention provides a method as defined above comprising coadministration, e.g., concomitantly or in sequence, of a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a second drug substance, said second drug substance being an anti-diabetic, a hypolipidemic agent, an anti-obesity agent or an anti-hypertensive agent, e.g., as indicated above.
Preferably, a compound of the invention is administered to a mammal in need thereof.
Preferably, a compound of the invention is used for the treatment of a disease which responds to modulation of the glucokinase activity.
Preferably, the condition associated with glucokinase activity is selected from impaired glucose tolerance, type 2 diabetes and obesity.
Finally, the present invention provides a method or use which comprises administering a compound of formula (I) in combination with a therapeutically effective amount of an antidiabetic agent, a hypolipidemic agent, an anti-obesity agent or an anti-hypertensive agent.
Ultimately, the present invention provides a method or use which comprises administering a compound of formula (I) in the form of a pharmaceutical composition as described herein.
As used throughout the specification and in the claims, the term "treatment" embraces all the different forms or modes of treatment as known to those of the pertinent art and in particular includes preventive, curative, delay of progression and palliative treatment.
The above-cited properties are demonstrable in vitro and in vivo tests using advantageously mammals, e.g., mice, rats, dogs, monkeys or isolated organs, tissues and preparations thereof. Said compounds can be applied in vitro in the form of solutions, e.g., preferably aqueous solutions, and in vivo either enterally, parenterally, advantageously intravenously, e.g., as a suspension or in aqueous solution. The dosage in vitro may range between about 10"2 molar and 10"9 molar concentrations. A therapeutically effective amount in vivo may range depending on the route of administration, between about 0.1 mg/kg and 1000 mg/kg, preferably between about 1 mg/kg and 100 mg/kg.
The activity of compounds according to the invention may be assessed by the following methods or methods well-described in the art: For example, the glucokinase activation in vitro may be determined by measuring the activation of recombinant GST-GK by a compound of the present invention in the absence or the presence of GKRP, a 68,000 Da protein inhibitor of GK. In these assays, formation of glucose-6-phosphate is coupled directly to the formation of thio-NADH. GST-GK catalyzes the reaction of glucose and Mg-ATP to produce glucose-6-phosphate and ADP. Glucose-6- phosphate dehydrogenase (G6PDH) reduces thionicotinamide (thio-NAD) to thio-NADH. The assay measures the formation of thio-NADH at 405 nM.
The basic GK assay components are as follows: 25 mM HEPES (pH 7.1), 25 mM KCI, 2.5 mM MgCI2, 1 mM ATP (Sigma A-5394), 1 mM DTT, 1 mM thio-NAD (Sigma T-7375), 80 units/mL G6PDH (Sigma G-5885), 10 mM glucose and 110 nM GST-GK. For assessing reversal of GK inhibition by GKRP, 20 μM Fructose-6-phosphate (F-6-P) and 370 nM recombinant GKRP are added to these assay components. Fructose-1 -phosphate (F-1-P) at 1 μM is used as a control in the GK/GKRP assay. F-1-P reverses inhibition of GST-GK by GKRP.
The assay is done in standard, 96-well, round-bottom plates (Corning) and the total assay volume is 25 μl_. Test compounds are serially diluted into 100% DMSO and 0.5 μl_ of diluted compound in 100% DMSO is added to the assay plate. Assay reagents (24.5 μl_) are added using a Zymark robotic platform. Buffer, containing HEPES, MgCI2, KCI, thio-NAD, G6PDH, glucose and GST-GK, are added (5 μL) using the Zymark 8-channel hand pipet. For the GK/GKRP assay, GKRP and F-6-P are also included. The reaction is then initiated by adding 19.5 μL of buffer containing HEPES, MgCI2, KCI, DTT and ATP using the Zymark Reagent Addition Station/Reagent Addition Module. The plates are read kinetically over 10 min at 25°C using a SpectraMax Plus microplate spectrophotometer (Molecular Devices, Sunnyvale, CA) to monitor the increase in optical density at 405 nm. The GK activity in wells containing test compounds is compared with activity in DMSO control wells. The concentration of compound that produces a 50% increase in the activity of GK is calculated and expressed as EC5O- A majority of the compounds described in the Examples had an EC50 value less than or equal to 200 μM and preferably less than 50 μM. Most preferable are compounds with EC50 less than 2μM which exhibited at least a 2-fold increase in % GK activation versus control.
The glucokinase activation in rat hepatocytes may be determined as follows: Hepatocytes are isolated by collagenase perfusion of the livers of overnight-fasted male Harlen Sprague-Dawley rats (Charles River Laboratories, Raleigh, NC) as previously described (see Berry et al., J. Cell Biol., Vol. 43, pp. 506-520 (1969)). The cells are washed three times each with 100 ml. of glucose-free Dulbecco's Modified Eagle medium (DMEM, Gibco BRL) containing 5% fetal bovine serum (FBS) and then suspended in glucose-free DMEM/5% FBS. Cells are plated in collagen coated 24-well plates (Becton Dickinson) at a density of 3 x 105 cells/well in 1 mL of William's Medium E (Sigma) supplemented with 5% FBS, and incubated at 37°C in 5% CO2/95% air. After cell attachment (~4 h), the medium is replaced with serum-free DMEM containing 5 mM glucose and 10 nM dexamethasone (Sigma), and cells are cultured further for 16-20 h prior to use.
The rate of glucose phosphorylation is determined by the release of 3H2O from [2-3H]glucose. The medium from the cultured hepatocytes is removed, and the cells are pre-incubated in 150 μL of fresh serum-free DMEM containing 5 mM glucose and compound (1 , 10 and 30 μM) or DMSO for 3 h at 370C. The final concentration of DMSO is 0.2%. The medium is then removed and 150 μL of a fresh mixture of DMEM/5 mM glucose containing compound or DMSO, and 1 μCi of [2-3H]glucose (NEN) is added. As a positive control for stimulation of glucose phosphorylation, cells are pre-incubated in serum-free DMEM/5 mM glucose medium containing DMSO for 3 h and then are incubated for 1 h in labeled glucose medium containing 0.5 mM fructose/DMSO (precursor of F-1-P, AnalaR® from BDH). All conditions are tested in quadruplicate where one well per plate received 200 μL of the appropriate medium plus labeled glucose (instead of 150 μL) of which 50 μL is immediately removed and placed in a 1.2 mL microfuge tube (Costar) containing 10 μL of 1 N HCI. This sample is used as a 0-minute time point for determining background 3H2O release (exchange values). Following the addition of the labeled glucose media, hepatocytes are incubated at 37°C on a slow moving rocker for 1 h.
On termination of the incubation, 50 μL of the culture medium is collected into microfuge tubes containing 10 μL of 1 N HCI, and determination of 3H2O. The tubes are left uncapped and each is placed inside a 20 mL glass scintillation vial (Wheaton) containing 1.5 mL of deionized water. The vials are capped tightly and incubated at 37°C in a dry incubator for 2 days (3H2O from the reaction mixture will equilibrate with the water in the vial). A standard curve is generated using [3H]H2O (NEN) to correct for exchange. 50 μL aliquots of serial dilutions of the labeled water are added to 10 μL of 1 N HCI and exchange is performed as described for the samples (typically, approximately 90% exchange is observed). The microfuge tubes are then removed from the vials carefully to minimize the removal of any water from the vial and 18 ml_ of scintillation cocktail (Ready Safe, Beckman Coulter) is then added to each vial. The 3H-label recovered from [2-3H]glucose in the water is determined using a Beckman Model LS500 scintillation counter and the counts (minus the 0-time point) are corrected for recovery of 3H2O. The amount of glucose de-tritiated in nanomoles/h per 106 cells is calculated, and the results are expressed as percent increase over the DMSO control.
The glucokinase activation in vivo may be determined as follows:
Male C57BL mice (Jackson Lab, Bar Harbor, ME) are housed 2 per cage in a reversed light cycle room (light on from 8:00 p.m. to 8:00 a.m.) and given access to food and water ad libitum. To induce DIO, the mice are given a high fat diet (D12492 with 60% caloric intake from fat, Research Diets, New Brunswick, NJ) from 4 weeks of age and maintained on the diet before being used. The DIO mice are used at 25 weeks of age. On the day of the study, animals are fasted at 7:30 a.m. Body weight measurement and basal blood sample collection are conducted at 10:00 a.m. Plasma glucose values are then determined. Animals are assigned into five groups (n=7/group) with the means of plasma glucose matched among the groups. At 10:30 a.m. animals are dosed with vehicle (water) or compound in vehicle with a dose volume of 5 ml/kg. The test compound is given at 3, 10, 30, or 100 mg/kg. One hour after vehicle or compound dosing, a blood sample (at 0 min) is taken followed by an oral glucose tolerance test (OGTT) at 1 g/kg (20% glucose in water) and a dose volume of 5 ml/kg. Blood samples are collected at 30, 60 and 120 min following the glucose administration. The animals are refed after the OGTT. Blood samples are taken via tail bleeding. Plasma glucose concentrations are determined using a glucose meter (Ascensia Elite, Bayer Corp., Mishawaka, IN). Blood samples are collected in tubes (Microvette CB300, Aktiengesellschaft & Co., Numbrecht, Germany) which contain EDTA (ethylene diaminetetraacetic acid) to prevent blood clotting. After blood sample collection, the tubes are kept on ice before being centrifuged. Plasma portion of the blood samples is obtained by centrifugation at 10,000 x g for 10 min at 4 0C and then stored at -80 0C. Plasma insulin levels are determined by Luminex assay using Mouse Endocrine Lincop/ex kit (Linco Research, Inc., St. Charles, MO). Data are reported as means ± SEM. Statistical analysis is performed using a one-way or two-way analysis of variance (ANOVA) followed by a Tukey post-hoc test to compare the difference among the groups. Statistical significance is accepted at the level of p<0.05. lllustrative of the invention, the compound of Example 1 demonstrates an EC50 of about 271 nM in the in vitro assay measuring the activation of recombinant GST-GK. Oral (po) dosing of a compound of Example 1 provides statistically significant glucose lowering with reductions in glucose of 38-53% at different timepoints during the oral glucose tolerance test.
The following Examples are intended to illustrate the invention and are not to be construed as being limitations thereon. If not mentioned otherwise, all evaporations are performed under reduced pressure, preferably between about 50 mmHg and 100 mmHg. The structure of final products, intermediates and starting materials is confirmed by standard analytical methods, e.g., microanalysis, melting point (m.p.) and spectroscopic characteristics, e.g., MS, IR and NMR. Abbreviations used are those conventional in the art.
Example 1
S-fBenzyl^-KRJ^-cyclopentyl-i^δ-methoxy-thiazolotδ^-blpyridin^-ylcarbamoyl)- ethyl]-benzenesulfonyl}-amino)-propionic acid
Figure imgf000035_0001
A. (4R,5S)-4-Methyl-5-phenyl-3-phenylacetyl-oxazolidin-2-one
(4R,5S)-4-Methyl-5-phenyl-oxazolidin-2-one (25.21 g, 142.269 mmol) is dissolved in anhydrous THF (100 ml_) and cooled to -780C. n-Butyl lithium (2.5 M in hexanes, 62.60 mL, 156.5 mmol) is added dropwise over 10 min. The reaction is stirred at -780C for 10 min. Phenylacetyl chloride (20.73 mL, 156.5 mmol) is dissolved in anhydrous THF (50 mL) and added dropwise over 10 min. The reaction is stirred at -780C for 2 h. The reaction is poured onto brine (300 mL) and extracted with ethyl acetate (2 x 250 mL). Extracts are combined, dried over anhydrous sodium sulfate, filtered, and concentrated to a yellow oil which solidified upon standing. Therude product is purified by column chromatography over silica eluting with 5→45% ethyl acetate in hexanes to afford (4R,5S)-4-methyl-5-phenyl-3- phenylacetyl-oxazolidin-2-one as a white solid: 1H NMR (400 MHz, DMSO-D6) δ 0.8 (d, J=6.6 Hz, 3 H) 4.2 (m, 2 H) 4.8 (m, 1 H) 5.9 (d, J=I. Z Hz, 1 H) 7.2 (m, 3 H) 7.3 (m, 5 H) 7.4 (m, 2H); LC/MS 296.2 (M+1), 294.1 (M-1).
B. Trifluoromethanesulfonic acid cyclopentylmethyl ester
Triflurormethanesulfonic anhydride (6.72 mL, 39.94 mmol) is dissolved in DCM (40 ml_) and cooled to -4O0C. To this is added dropwise cyclopentanemethanol (4.00 g, 39.94 mmol) dissolved in DCM (10 mL) and anhydrous pyridine (3.23 mL, 39.94 mmol). The reaction is stirred at -4O0C for 45 min. The reaction is diluted with hexanes (30 mL) and run through a plug of silica using hexanes (300 mL) as the eluent. Concentration without heating gives trifluoromethanesulfonic acid cyclopentylmethyl ester as a clear, colorless oil that is used immediately.
C. (4R,5S)-3-((R)-3-Cyclopentyl-2-phenyl-propionyl)-4-methyl-5-phenyl-oxazolidin-2- one
The title A compound, (4R,5S)-4-methyl-5-phenyl-3-phenylacetyl-oxazolidin-2-one (5.00 g, 16.93 mmol) is dissolved in anhydrous THF (50 mL) and cooled to -780C. Lithium bis(trimethylsilyl)amide (1 M in THF, 16.93 mL, 16.93 mmol) is added dropwise over 10 min. To this is added a solution of the title B compound, trifluoromethanesulfonic acid cyclopentylmethyl ester (3.93 g, 16.93 mmol) in anhydrous THF (15 mL). The reaction is stirred at -780C for 10 min, then the cold bath is removed. The reaction is allowed to stir and warm to RT. After one hour, the reaction is concentrated to remove the solvent, resulting in a yellow oil. This is dissolved in ethyl acetate (300 mL) and washed with water (2 x 200 mL). The organic solution is dried over anhydrous sodium sulfate, filtered, and concentrated to a yellow oil. This is recrystallized from hexanes to afford (4R,5S)-3-((R)-3-cyclopentyl-2- phenyl-propionyl)-4-methyl-5-phenyl-oxazolidin-2-one as a white solid: 1H NMR (400 MHz, DMSO-D6) δ 0.8 (d, J=6.6 Hz, 3 H) 1.1 (d, J=16.2 Hz, 2 H) 1.4 (m, 2 H) 1.5 (d, J=5.8 Hz, 2 H) 1.6 (s, 2 H) 1.8 (d, J=7.1 Hz, 2 H) 2.0 (d, J=13.4 Hz, 1 H) 4.8 (s, 1 H) 5.1 (s, 1 H) 5.7 (d, J=7.1 Hz, 1 H) 7.3 (s, 1 H) 7.4 (m, 9 H); LC/MS 378.2 (M+1).
D. (R)-3-Cyclopentyl-2-phenyl-propionic acid
Lithium hydroxide monohydrate (1.28 g , 30.6 mmol) is suspended in THF (50 mL) and cooled to O0C. Hydrogen peroxide (50 wt% in water, 2.81 mL, 91.79 mmol) is added dropwise. Reaction is stirred at O0C for 15 min and the title C compound, (4R,5S)-3-((R)-3- cyclopentyl-2-phenyl-propionyl)-4-methyl-5-phenyl-oxazolidin-2-one (3.85 g, 10.2 mmol) is added in five separate portions, allowing portions to dissolve completely between additions. The reaction is allowed to stir and warm to RT overnight. The reaction is then concentrated to a sticky white solid. This is dissolved in ethyl acetate (25 ml_)/1 N HCI (25 ml_). The organic solution is dried over anhydrous sodium sulfate, filtered, and concentrated to afford the crude product as a yellow oil. The crude product is chromatographed over silica eluting with 5->65% ethyl acetate in hexanes to afford (R)-3-cyclopentyl-2-phenyl-propionic acid as a white solid: 1H NMR (400 MHz, CDCI3) δ 1.1 (m, J=11.6, 7.8, 7.8, 3.9 Hz, 2 H) 1.5 (m, 2 H) 1.6 (m, 2 H) 1.7; LC/MS 217.2 (M-1).
E. 5-Methoxy-thiazolo[5,4-b]pyridin-2-ylamine
Potassium thiocyanate (156 g, 1600 mmol) is dissolved in acetic acid (1400 ml.) and cooled to O0C. 6-Bromo-pyridin-3-ylamine (50 g, 400 mmol) is dissolved in acetic acid (100 mL) and added dropwise over 10 min. Bromine (25 mL, 480 mmol) is dissolved in acetic acid (100 mL) and added dropwise over 10 min. The reaction is allowed to stir and warm to RT overnight. Acetic acid is removed via concentration. The resulting residue is poured into water (1 L) and adjusted to pH = 7 with 1 N sodium hydroxide solution. This is extracted with ethyl acetate. Extracts are combined, washed with brine, and concentrated to a brown sludge. Recrystallization from methanol affords 5-methoxy-thiazolo[5,4-b]pyridin-2-ylamine as a brown solid: 1H NMR (400 MHz, DMSO-D6) δ 3.8 (s, 3 H) 6.7 (d, J=8.8 Hz, 1 H) 7.4 (s, 2 H) 7.6 (d, J=8.6 Hz, 1 H); LC/MS 182.3 (M+1), 180.4 (M-1).
F. (R)-3-Cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-phenyl- propionamide
The title D compound, (R)-3-cyclopentyl-2-phenyl-propionic acid (1.66 g, 7.605 mmol) is dissolved in thionyl chloride (15 mL) and heated at 5O0C for 1.5 h. The reaction is then cooled to RT and concentrated to afford a yellow oil. This is dissolved in anhydrous THF (10 mL) and added dropwise to a solution of the title E compound, 5-methoxy-thiazolo[5,4- b]pyridin-2-ylamine (1.38 g, 7.605 mmol) in anhydrous pyridine (10 mL) at O0C. The reaction is allowed to stir and warm to RT. After 2 h, the reaction is concentrated to remove the THF and most of the pyridine. The resulting brown oil is dissolved in ethyl acetate (250 mL)/1 N HCI (250 mL) and extracted further with ethyl acetate. The combined organic layer is dried over anhydrous sodium sulfate, filtered, and concentrated to afford the crude product as a brown oil. This is chromatographed over silica eluting with 5→60% ethyl acetate in hexanes to afford (R)-3-cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-phenyl-propionamide as a yellow foam: 1H NMR (400 MHz, DMSO-D6) δ 1.1 (m, 2 H) 1.4 (dd, J=7.2, 4.7 Hz, 2 H) 1.6 (m, 3 H) 1.7 (m, 3 H) 2.1 (m, i H) 3.9 (m, 4 H) 6.9 (d, J=8.8 Hz, 1 H) 7.3 (m, 1 H) 7.4 (m,
4 H) 8.0 (d, J=8.6 Hz, 1 H) 12.5 (s, 1 H); LC/MS 382.1 (M+1), 380.2 (M-1).
G. 4-[(R)-2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-/j]pyridin-2-ylcarbamoyl)-ethyl] benzenesulfonyl chloride
Chlorosulfonic acid (20 ml_) is cooled to O0C. To this is added a solution of the title F compound, (R)-3-cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-phenyl- propionamide (2.53 g, 6.632 mmol) in DCM (10 ml_). The reaction is allowed to stir and warm to RT over 2.5 h. The reaction is then added dropwise to crushed ice (400 ml.) and extracted with DCM (3 x 100 ml_). Extracts are combined, dried over anhydrous sodium sulfate, filtered, and concentrated to give 4-[(R)-2-cyclopentyl-1-(5-methoxy-thiazolo[5,4- 6]pyridin-2-ylcarbamoyl)-ethyl] benzenesulfonyl chloride as a yellow foam. Product is isolated as a 80:20 mixture of the desired para product and undesired meta product: 1H NMR (400 MHz, DMSO-D6) δ 1.1 (s, 2 H) 1.4 (d, J=4.8 Hz, 1 H) 1.4 (s, 1 H) 1.5 (m, 3 H) 1.7 (s, 2 H) 1.8 (d, J=13.4 Hz, 1 H) 2.1 (m, 1 H) 3.9 (s, 3 H) 4.0 (s, 1 H) 6.9 (d, J=8.6 Hz, 1 H) 7.3 (m, 2 H) 7.6 (d, J=8.1 Hz, 2 H) 8.0 (d, J=8.6 Hz, 1 H) 14.3 (s, 2 H); LC/MS 480.2 (M+1), 478.3 (M-1).
H. 3-benzyl amino propionic acid tert butyl ester
Benzylamine (1 g, 9.33 mmol) is dissolved in ethanol (25 ml_), to which tert-butyl acrylate (1.20 g, 9.33 mmol) is added. The reaction is stirred at RT overnight and then concentrated to a crude yellow oil. This material is purified via flash chromatography (2-»20% methanol in ethyl acetate) to afford 3-benzyl amino propionic acid tert butyl ester as a clear oil: 1H NMR (400MHz, CDCI3) δ 1.44 (s, 9H) 1.64 (s, 1 H) 2.44 (m, 2H) 2.85 (m, 2H) 3.79 (m, 2H) 7.24 (m, 1 H) 7.31 (m, 4H).
I. S^Benzyl^-KRJ^-cyclopentyl-i^δ-methoxy-thiazolotδ^-Jblpyridin-Z- ylcarbamoyl)-ethyl]-benzenesulfonyl}-amino)~propionic acid tert-butyl ester
The title G compound, 4-[(R)-2-cyclopentyl-1-(5-methoxythiazole[5,-4-ib]pyridin-2- ylcarbomyl)ethyl] benzene sulfonyl chloride (0.301 g, 0.627 mmol) is dissolved in DCM (10 ml_), to which the title H compound, 3 benzyl amino propionic acid tert butyl ester (0.148 g, 0.627 mmol) and DIEA (0.162 g, 1.254 mmol) in DCM are added. The reaction mixture is stirred at RT for one hour and then concentrated. The residue is partitioned between 1 N HCI / ethyl acetate. The organic layer is dried over magnesium sulfate, filtered and concentrated to afford a crude yellow oil. This material is purified via flash chromatography (10% ethyl acetate in hexanes → ethyl acetate) to afford a pale yellow foam. This material is separated from the meta isomer (derived from compound G) by chiral HPLC (20% ethanol in hexanes) to afford 3-(benzyl-{4-[(R)-2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2- ylcarbamoyl)-ethyl]-benzenesulfonyl}-amino)-propionic acid tert-butyl ester as a white foam: 1H NMR (400MHz, CDCI3) δ 1.16 (m, 2H) 1.35 (s, 9H) 1.51 (m, 2H) 1.64 (m, 3H) 1.76 (m, 2H) 1.96 (m, 1 H) 2.26 (m, 1 H) 2.36 (m, 2H) 3.40 (m, 2H) 3.69 (m, 1 H) 4.00 (s, 3H) 4.36 (s, 2H) 6.81 (m, 1 H) 7.28 (m, 5H) 7.49 (m, 2H) 7.82 (m, 3H) 8.65 (s, 1 H); LC/MS 679.6 (M+1), 677.6 (M-1).
J. 3-(BenzyI-{4-[(R)-2-cyclopentyl-1-(5-methoxy-thia2olo[5,4-Jb]pyridin-2- ylcarbamoyl)-ethyl]-benzenesulfonyl}-amino)-propionic acid
The title I compound, 3-(benzyl-{4-[(R)-2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-jb]pyridin-2- ylcarbamoyl)-ethyl]-benzenesulfonyl}-amino)-propionic acid tert-butyl ester (0.064 g, 0.0942 mmol) is dissolved in methanol (5 ml_), to which a solution of sodium hydroxide (0.004 g, 0.0942 mmol) in water (2 ml.) is added. The reaction mixture is stirred at RT overnight. The reaction mixture is then concentrated and the residue is partitioned between 1 M HCI / ethyl acetate. The organic layer is dried over magnesium sulfate, filtered and concentrated to afford 3-(benzyl-{4-[(R)-2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-ιb]pyridin-2-ylcarbamoyl)- ethyl]-benzenesulfonyl}-amino)-propionic acid as a white foam: 1H NMR (400MHz - CDCI3) δ 1.17 (m, 2H) 1.50 (m, 2H) 1.66 (m, 4H) 1.78 (m, 1 H) 2.28 (m, 1 H) 2.43 (m, 2H) 3.23 (m, 1 H) 3.50 (m, 1 H) 3.85 (m, 2H) 4.00 (s, 3H) 4.37 (m, 1 H) 4.77 (m, 1 H) 6.79 (m, 1 H) 7.36 (m, 5H) 7.52 (m, 2H) 7.67 (m, 1 H) 7.95 (m, 2H); LC/MS 623.3 (M+1), 621.4 (M-1). EC50 in primary enzyme assay 0.27μM
Example 2
3-[{4-[2-Cyclopentyl-1-([1,3,4]thiadiazol-2-ylcarbamoyl)-ethyl]-benzene-sulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid
Figure imgf000040_0001
A. S-Cyclopentyl^-phenylpropionic acid methyl ester.
To a 500 mL round bottom flask containing 250 ml_ of 9:1 THF/DMPU at -780C is added 17 mL DIEA followed by rapid addition of 49 mL of 2.5 M n-BuLi/hexanes. After 10 min at - 780C, a solution of ethyl phenylacetate (17.0 g,113 mmol) in 50 mL of 9:1 THF/DMPU is added dropwise over 15-20 min. A yellow solution results, and the reaction mixture is stirred at -780C for 30-45 min. A solution of 26.7 g (115 mmol) of freshly prepared cyclopentyl triflate (title B compound in Example 1) in 25 mL 9:1 THF/DMPU is added dropwise over 10- 15 min. The mixture is then stirred over 2-3 h to ambient temperature. The reaction is quenched into 300 mL of 1 N HCI and extracted 3 times with methyl-t-butylether (MTBE). The combined extracts are washed with saturated NaCI and dried over anhydrous magnesium sulfate. Filtration and evaporation afford crude product as a yellow oil. Flash chromatography over silica gel eluting with 4:1 hexane/MTBE affords 3-cyclopentyl-2- phenylpropionic acid methyl ester as a light yellow oil: 1H NMR (400 MHz, CDCI3) δ 1.1 (m, 2H), 1.3 (m, 1 H), 1.50 (m, 2H), 1.6 (m, 2H), 1.7 (m, 2H), 2.1 (dt, J =13.4, 7.7 Hz, 1 H), 3.60 (m, 1 H), 3.64 (s, 3H), 7.3 (m, 5H); LC/MS 233 (M+1).
B. S-Cyclopentyl^-phenylpropionic acid.
To title A compound, S-cyclopentyl^-phenylpropionic methyl ester (13.0 g, 56 mmol) in 100 mL of methanol is added a solution of 5.6 g (140 mmol) of NaOH in 8 mL of water. The reaction mixture is stirred at ambient temperature overnight. The methanol is evaporated and the residue is dissolved in 200 mL of water and extracted with ethyl ether. The aqueous phase is separated and adjusted to pH 2-3 with 1 N HCI and extracted with MTBE twice. The combined extracts are washed with saturated sodium chloride and dried over anhydrous magnesium sulfate. The material is evaporated to a yellow oil that crystallizes to provide 3- cyclopentyl-2-phenylpropionic acid as an off-white solid: 1H NMR (400 MHz, CDCI3) δ 1.7 (m, 2H), 2.1 (m, 2H), 2.2 (m, 3H), 2.4(m, 3H), 2.4 (m,1H), 4.2 (t, J=7.7 Hz, 1H), 7.9 (m, 5H); LC/MS 236 (M+NH4 +).
C. S-Cyclopentyl-^-phenylpropionyl chloride
To the title B compound, S-cyclopentyl^-phenylpropionic acid (3.1 g,14.2 mmol) is added 10 ml. of thionyl chloride and one drop of DMF. The reaction mixture is stirred at ambient temperature for 2 h. The excess thionyl chloride is evaporated and the residue is twice treated with toluene and evaporated to yield 3-cyclopentyl-2-phenylpropionyl chloride as an orange-yellow oil which is sed as such without further purification.
D. 3-Cyclopentyl-2-phenyl-N-[1,3,4]thiadiazol-2-yl-propionamide
To 2-amino-1 ,3,4-thiadiazole (220 mg, 21 mmol), 0.4 mL DIEA and 7 ml_ of DCM is added a solution of the title C compound, S-cyclopentyl^-phenylpropionyl chloride (500 mg, 21 mmol) in 2 mL of DCM. The mixture is stirred at ambient temperature for 2-3 h, the mixture is evaporated, and the residue is treated with 10 mL 1 N HCI and extracted twice with ethyl acetate. The combined extracts are washed with brine, saturated sodium bicarbonate solution, and brine and dried over anhydrous magnesium sulfate. Filtration and evaporation afford 3-cyclopentyl-2-phenyl-N-[1 ,3,4]thiadiazol-2-yl-propionamide as an orange foam: 1H NMR (400 MHz, DMSO-d6) δ 11 (m, 2H), 1.4 (m, 2H), 1.5 (m, 3H), 1.7(m, 3H), 2.1 (ddd, J=13.3, 7.8, 7.7 Hz, 1 H), 4.0 (dd, J=8.1 , 7.1 Hz, 1 H), 7.2 (s, 1 H), 7.3 (m, 4H), 9.1 (s, 1H), 12.8 (s, 1H); LC/MS 302 (M+1), 300 (M-1).
E. 4-[2-Cyclopentyl-1 -(1 ,3,4)thiadiazol-2-ylcarbamoyl)-ethyl]benzenesulfonyl chloride
To 5 mL of chlorosulfonic acid in an ice bath at O0C is added dropwise title D compound, 3- cyclopentyl-2-phenyl-N-[1 ,3,4]thiadiazol-2-yl-propionamide (200 mg, 0.66 mmol) in 2 mL of DCM. The reaction is stirred at ambient temperature for 2-3 h until an orange color persists. The reaction is carefully quenched into solid ice, extracted with ethyl acetate and dried over anhydrous magnesium sulfate. The reaction is filtered and reduce to a greenish resin which is used as such in the next step.
F. 3-[{4-[2-Cyclopentyl-1 -([1 ,3,4]thiadiazol-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}- (2-methoxy-ethyl)-amino]-propionic acid tert-butyl ester
To t-butyl N-(2-methoxyethyl)propionate (110 mg, 0.058 mmol) and 0.20 mL of DIEA in 8 mL of DCM is added title E compound, 4-[2-cyclopentyl-1-(1 ,3,4)thiadiazol-2-ylcarbamoyl)- ethyl]benzenesulfonyl chloride (230 mg, 0.57 mmol). The mixture is stirred at ambient temperature for one hour and DCM is evaporated. The residue is treated with 10 mL of 1 N HCI and extracted with ethyl acetate. The organic extract is washed successively with brine, saturated sodium bicarbonate, and brine. The organic layer is dried over anhydrous magnesium sulfate, filtered and evaporated to a brown oil. This is purified using a Biotage 40M column eluted with 1 :1-hexane:ethyl acetate to provide 3-[{4-[2-cyclopentyl-1- ([1 ,3,4]thiadiazol-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid tert-butyl ester as a light orange foam: 1H NMR (400 MHz, DMSOd6) δ 1.1 (m, 1H), 1.4 m, 5H), 1.5 (m, 1H), 1.7 (m, 1 H), 2.4 (t, 7.3 Hz, 1 H), 3.3 (t, J=5.7 Hz, 1 H), 3.3 (m, 5H), 7.61 (d, J=8.3 Hz, 2H), 7.79 (d, J=8.3 Hz, 2H), 9.16 (s, 1 H), 12.89 (s, 1 H); LC/MS 567 (M+1), 565 (M-1).
G. 3-[{4-[2-Cyclopentyl-1 -([1 ,3,4]thiadiazol-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}- (2-methoxy-ethyl)-amino]-propionic acid
To 75 mg (0.133 mmol) of title F compound, 3-[{4-[2-cyclopentyl-1-([1 ,3,4]thiadiazol-2- ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid tert-butyl ester is added 2 mL of DCM and 0.5 mL of trifluoroacetic acid. The reaction mixture is stirred at ambient temperature for one hour. The reaction is evaporated to a brownish resin and subsequently dissolved in ether and decanted from some insoluble material. The organic layer is evaporated to a light orange resin which is dissolved in 2 mL methanol in a 50 mL round bottom flask. Aqueous potassium hydroxide (0.265 mL of 0.5094 M potassium hydroxide solution) is added and stirred for 30 min. The methanol is evaporated, 8 mL of water added, and the mixture lyophilized to afford 3-[{4-[2-cyclopentyl-1-([1 ,3,4]thiadiazol-2- ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid as a light orange-brown solid: 1H NMR (400 MHz, DMSO-d6) δ 1.1 (m, 1 H), 1.4 (m, 1H), 1.6 (m, 2H), 1.7 (ddd, J=13.1 , 6.9, 6.7 Hz, 2H), 2.1 (ddd, J=11.2, 8.2, 7.8, 2H), 3.2 (s, 2H), 3.2 (m, 3H), 3.7 (t, J=7.5 Hz, 1 H), 7.56 (d, J=8.3, 2H), 7.63 (d, J=8.3 Hz, 2H), 8.58 (s, 1 H); L/MS 511 (M+1), 509 (M-1). EC50 in primary enzyme assay 50μM.
Example 3
S-tBenzofuran^-ylmethyl^-KRJ^-cyclopentyl-i-tδ-methoxy-thiazolotδ^-JbJpyridin^- ylcarbamoyl)-ethyl]-benzenesulfonyl}-amino)-propionic acid
Figure imgf000043_0001
A. 3-Cyclopentyl-2-(4-nitro-phenyl)-propionic acid ethyl ester
To a 1 L round bottom flask containing 250 mL of 9:1 THF/DMPU at -78C are added under nitrogen 11 mL (78.6 mmol) anhydrous DIEA followed by rapid addition of 32 mL of 2.5 M n- BuLi in hexanes. After 10 min at -780C a solution of 15.4 g (74 mmol) of p-nitrophenylacetic acid, ethyl ester in 100 mL of 9:1 THF/DMPU is added dropwise over 30 min. A deep purple solution results, and the reaction mixture is stirred at -780C for 30 min and then cyclopentyl methyl iodide (17.6 g, 78 mmol) in 50 mL of 9:1 THF/DMPU is added. The reaction is stirred while warming slowly to RT overnight. The mixture is poured into 1 L of 1 N HCI and extracted twice with MTBE. The combined MTBE extracts are washed with brine, dried over anhydrous magnesium sulfate, filtered and reduced to an orange oil. Flash chromatography over silica eluting with 4:1 hexane/MTBE affords 3-cyclopentyl-2-(4-nitro-phenyl)-propionic acid ethyl ester as an orange oil: 1H NMR (400 MHz, CDCI3) δ 1.0-1.1 (m, 2H), 1.2 (t, 3H, J=7.2), 1.4-1.8 (m, 5H), 1.8-1.9 (m, 2H), 2.1-2.25 (m, 2H), 3.74 (t, 1 H, J=7.8), 4.1 (m, 2H), 7.51 (d, 2H, J=8.8), 8.19 (d, 2H, J=8.8); LC/MS 290 (M-1).
B. 3-Cyclopentyl-2-(4-nitro-phenyl)-propionic acid
The title A compound , 3-cyclopentyl-2-(4-nitro-phenyl)-propionic acid ethyl ester (3.6 g, 12.3 mmol) is dissolved in 25 mL of methanol and aqueous NaOH (0.70 g, 17.5 mmol in 4 mL of water) is added and the mixture is stirred at RT overnight. The methanol is removed under reduced pressure and the residue is diluted with 100 mL of water and extracted with ether. The aqueous layer is then acidified with 1 N HCI and then extracted with ethyl acetate. The combined ethyl acetate layers are dried over anhydrous magnesium sulfate, filtered and reduced under vacuum to a crude orange oil. The crude oil is triturated with 100 mL of hexane/10-15mL of ether to produce 3-cyclopentyl-2-(4-nitro-phenyl)-propionic acid as a solid: 1H NMR (400 MHz, CDCI3) δ 1.0-1.1 (m, 2H), 1.4-1.8 (m, 5H), 1.8-1.9 (m, 2H), 2.1-2.25 (m, 2H), 3.74 (t, 1 H1 J=7.8), 7.51 (d, 2H, J=8.8), 8.19 (d, 2H1 J=8.8); LC/MS 218 (-CO2, M-1), 279 (M+NH4 +).
C. 3-Cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-(4-nitro-phenyl)- propionamide
The title B compound, 3-cyclopentyl-2-(4-nitro-phenyl)-propionic acid (7.5 g, 28.5 mmol) is dissolved in 25 mL of thionyl chloride and a drop of DMF and the mixture stirred at RT for 5- 6 h. The excess of thionyl chloride is removed under reduced pressure. The residue is then taken up in DCM and added dropwise to a solution of the title E compound in Example 1 , 5-methoxy-thiazolo[5,4-b]pyridin-2-ylamine (5.2 g, 28.5 mmol) in 25 mL of pyridine. The reaction mixture is stirred for 5 h before being evaporated to remove the pyridine. The residue is partitioned between ethyl acetate and brine, extracted with ethyl acetate. The combined organic layers are washed with saturated sodium bicarbonate, brine, dried over anhydrous magnesium sulfate, filtered and then reduced to an orange-brown solid. This is then vacuum dried to afford 3-cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-(4- nitro-phenyl)-propionamide as a foam: 1H NMR (400 MHz, CDCI3) δ 1.0-1.1 (m, 2H)1 1.4-1.8 (m, 5H), 1.8-1.9 (m, 2H)1 2.1-2.25 (m, 2H), 3.6 (t, 1 H, J=7.8), 4.01 (s, 3H), 6.8 (d, 1 H, J=8.8), 7.4 (d, 2H, J=8.6), 7.8 (d, 1H, J=8.8 Hz), 8.19 (d, 2H, J=8.6 Hz), 9.3 (s, 1 H); LC/MS 427 (M+1).
D. 2-(4-Amino-phenyl)-3-cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)- propionamide
The title C compound, 3-cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin-2-yl)-2-(4-nitro- phenyl)-propionamide (12 g, 28.2 mmol) is diluted with 160 mL of ethanol and 150 mL acetic acid. 8 g of iron powder (325 mesh, 0.14 mol) is added and the mixture heated to reflux. Once reflux begins the mixture is stirred vigorously and then heating is discontinued and the mixture is allowed to cool slowly. The solvents are removed and the residue is treated with 250 mL of water. Saturated sodium bicarbonate ia added carefully to bring the mixture to a pH of 8-9. The mixture is extracted with ethyl acetate, washed with brine, dried and evaporated to give an orange solid which is triturated from hexane. The resulting solid is collected by filtration to afford 2-(4-amino-phenyl)-3-cyclopentyl-N-(5-methoxy-thiazolo[5,4- b]pyridin-2-yl)-propionamide: 1H NMR (400 MHz1 CDCI3) δ 1.0-1:1 (m, 2H)1 1.4-1.8 <m, 5H)1 1.8-1.9 (m, 2H)1 2.1-2.25 (m, 2H), 3.6 (t, 1 H, J=7.8), 3.98 (s, 3H), 6.7 (d, 1H1 J=8.8), 6.8 (d, 2H, J=8.6), 7.2 (d, 2H, J=8.6), 7.8 (d, 1 H, J=8.8); LC/MS 397 (M+1). E. 4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)"ethyl]- benzenesulfonyl chloride
The title D compound, 2-(4-amino-phenyI)-3-cyclopentyl-N-(5-methoxy-thiazolo[5,4-b]pyridin- 2-yl)-propionamide (2.0 g, 5.1 mmol) is dissolved in 50 mL of acetic acid and 20 ml_ of concentrated HCI and the mixture is cooled to O0C. A solution of 0.35 g (5.1 mmol) of NaNO2 in 5 mL of water is added dropwise and the mixture is stirred for 30 min. The resulting yellow solution is then added to 180 mL of the Green Solution (prepared by bubbling 74 g of sulfur dioxide gas into 740 mL of glacial acetic acid followed by addition of 30 g of CuCb in 35-40 mL water. The resulting mixture is filtered through filter paper to obtain a clear green solution) and the mixture is stirred at RT overnight (the initial black- green solution transforms to a light green solution after 24 h). The resulting mixture is poured onto 500 g of ice and the precipitated solids are collected by filtration, washed with water and then dissolved in ethyl acetate, washed with brine, dried over anhydrous magnesium sulfate, filtered and evaporated to afford a yellow foam. This material is flash chromatographed over silica eluting with 7:3 hexane/ethyl acetate to afford 4-[2-cyclopentyl- 1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]-benzenesulfonyl chloride as a stable yellow foam: 1H NMR (400 MHz, CDCI3) δ 1.0-1.1 (m, 2H), 1.4-1.8 (m, 5H), 1.8-1.9 (m, 2H), 2.1-2.25 (m, 2H), 3.7 (t, 1H, J=7.8), 4.01 (s, 3H), 6.8 (d, 1 H, J=8.8), 7.5 (d, 2H, J=8.6), 7.8 (d, 1H, J=8.8), 8.19 (d, 2H, J=8.6), 9.3 (s, 1H); LC/MS 480 (M+1).
F. 2-Benzofuran-2-ylmethyl-isoindole-1 ,3-dione
Benzofuran-2-yl-methanol (450 mg, 3.037 mmol) is dissolved in THF (100 mL). Triphenylphosphine (956 mg, 3.645 mmol) and phthalamide (536 mg, 3.645 mmol) are added in one portion. The reaction is cooled to 00C. Di-tert-butyl azodicarboxylate (839 mg, 3.645 mmol) is dissolved in THF (10 mL) and added dropwise. The reaction is allowed to warm to RT overnight and the solvent is removed by rotary evaporation. The resulting orange oil is triturated in methanol and filtered to afford 2-benzofuran-2-ylmethyl-isoindole- 1 ,3-dione as a white solid: 1H NMR (400 MHz, DMSO-D6) δ 4.9 (s, 2 H) 6.87 (s, 1 H) 7.2 (m, 2 H) 7.5 (m, 2 H) 7.9 (m, 4 H); LC/MS 278.1 (M+1).
G. Benzofuran-2-yI-methylamine
The title F compound, 2-benzofuran-2-ylmethyl-isoindole-1 ,3-dione (1 g, 3.606 mmol) is slurried in methanol (150 mL). Hydrazine hydrate (271 mg, 5.410 mmol) is added and the reaction heated at 500C for 2.5 h. A solution is formed during heating. The reaction is then cooled to RT and concentrated to afford a white foam. This is dissolved in 1 N sodium hydroxide solution and ethyl acetate. The organic layer is separated, dried over anhydrous sodium sulfate, filtered, and concentrated to afford benzofuran-2-yl-methylamine as a yellow solid: 1H NMR (400 MHz, CDCI3) δ 4.0 (s, 2 H) 6.5 (s, 1 H) 7.2 (m, 2 H) 7.4 (d, J=7.8 Hz, 1 H) 7.5 (d, J=7.6 Hz, 1 H); LC/MS 131.1 (M+1).
H. 3-[(Benzofuran-2-ylmethyl)-amino]-propionic acid benzyl ester
The title G compound, benzofuran-2-yl-methylamine (290 mg, 1.970 mmol) is dissolved in ethanol (50 ml_). To this is added benzyl acrylate (352 mg, 2.167 mmol). The reaction is stirred at RT overnight, then concentrated to afford the crude product as a yellow oil. This is chromatographed using 20% ethyl acetate in hexanes → ethyl acetate to afford the purified product as a pale yellow oil: 1H NMR (400 MHz, CDCI3) δ 2.6 (t, J=6.4 Hz, 2 H) 3.0 (t, J=6.4 Hz, 2 H) 3.9 (s, 2 H) 5.1 (s, 2 H) 6.5 (d, J=1.0 Hz, 1 H) 7.2 (m, 2 H) 7.3 (m, 5 H) 7.4 (m, 1 H) 7.5 (dd, J=7.5, 1.1 Hz, 1 H); LC/MS 310.1 (M+1).
I. 3-(Benzofuran-2-ylmethyl-{4-[(R)-2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin- 2-ylcarbamoyl)-ethyl]-benzenesuIfonyl}-amino)-propionic acid benzyl ester
The title E compound, 4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)- ethyl]-benzenesulfonyl chloride (1.208 mg, 2.517 mmol) is added in one portion to a solution of the title H compound, 3-[(benzofuran-2-ylmethyl)-amino]-propionic acid benzyl ester (856 mg, 2.768 mmol) and TEA (388 μl_, 2.768 mmol) in DCM, and stirred at RT overnight. The reaction is concentrated to afford a green oil. Purification via column chromatography eluting with10% ethyl acetate in hexanes -» ethyl acetate afforded the racemic product as a tan solid (750 mg). This is separated by chiral prep HPLC to afford the desired steroisomer of 3-(benzofuran-2-ylmethyl-{4-[(R)-2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2- ylcarbamoyl)-ethyl]-benzenesulfonyl}-amino)-propionic acid benzyl ester as a white solid: 1H NMR (400 MHz, DMSO-D6) δ 1.1 (m, 2 H) 1.4 (dd, J=7.2, 4.7 Hz, 2 H) 1.6 (dt, J=14.9, 7.5 Hz, 3 H) 1.7 (ddd, J=13.4, 7.1 , 6.8 Hz, 3 H) 2.1 (ddd, J=13.1 , 7.8, 7.6 Hz, 1 H) 2.6 (m, 2 H) 3.4 (m, 2 H) 3.9 (s, 3 H) 4.0 (t, J=7.6 Hz, 1 H) 4.6 (d, J=11.4 Hz, 2 H) 5.0 (s, 2 H) 6.7 (s, 1 H) 6.9 (m, 1 H) 7.1 (m, 2 H) 7.3 (m, 6 H) 7.5 (m, 1 H) 7.6 (d, J=8.3 Hz, 2 H) 7.8 (d, J=8.3 Hz, 2 H) 8.0 (d, J=8.8 Hz, 1 H) 12.7 (s, 1 H); LC/MS 753.6 (M+1), 751.7 (M-1).
J. 3-(Benzofuran-2-ylmethyl-{4-[(R)-2-cyclopentyl-1-(5-methoxy-thiazolo[5,4- blpyridin^-ylcarbamoyO-ethyll-benzenesulfony^-amino^propionic acid The title I compound, 3-(benzofuran-2-ylmethyl-{4-[(R)-2-cyclopentyl-1-(5-methoxy- thiazolo[5,4-ϋ]pyridin-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-amino)-propionic acid benzyl ester (280 mg, 0.372 mmol) is dissolved in THF. To this is added a solution of sodium hydroxide (22 mg, 0.558 mmol) in water. The reaction is stirred at RT overnight. LC/MS indicates ~80% reaction completion and, therefore, more sodium hydroxide (10 mg, 0.250 mmol) is added. After 2 h of stirring at RT, the reaction is acidified to pH 1 with concentrated hydrochloric acid, then concentrated to remove solvent. The residue is diluted with ethyl acetate and water. The organic layer is separated, dried over anhydrous sodium sulfate, filtered, and concentrated to afford the crude product as a yellow foam. Purifiaction by preparative HPLC affords 3-(benzofuran-2-ylmethyl-{4-[(R)-2-cyclopentyl-1-(5-methoxy- thiazolotδ^-όjpyridin^-ylcarbamoylj-ethyll-benzenesulfonylj-aminoj-propionic acid as a white solid: 1H NMR (400 MHz, DMSO-D6) δ 1.4 (m, 2 H) 1.6 (m, 3 H) 1.7 (m, 3 H) 2.1 (m, 1 H) 3.35 (m, 1 H) 3.9 (s, 3 H) 4.0 (m, 1 H) 4.6 (s, 2 H) 6.7 (s, 1 H) 6.9 (d, J=8.8 Hz, 1 H) 7.1 (m, 2 H) 7.3 (m, 2 H) 7.5 (dt, J=4.3, 2.1 Hz, 1 H) 7.6 (d, J=8.3 Hz, 2 H) 7.8 (d, J=8.6 Hz, 2 H) 8.0 (d, J=8.8 Hz, 1 H) 12.7 (s, 1 H); LC/MS 663.5 (M+1), 661.5 (M-1). EC50 in primary enzyme assay 0.22μM.
Example 4
The following examples may be prepared by a skilled artisan using the appropriated methods described herein above.
4-1 3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-pyridin-2-ylmethyl-amino)-propionic acid: MS MH+ 624; 1 H NMR (400 MHz, DMSO-D6) δ ppm 1.04 1.2 (m, 2 H) 1.40 - 1.48 (m, 2 H) 1.52 - 1.62 (m, 3 H) 1.68 - 1.75 (m, 2 H) 1.78 - 1.85 (dd, J=13.5, 7.2 Hz, 1 H) 2.10 - 2.20 (m, 1H) 2.4 (m, 2 H) 3.4 (t, J=7.5 Hz, 2 H) 3.91 (s, 3H) 4.1 (t, J=7.5 Hz, 1 H) 4.5 (s, 2 H) 6.9 (d, J=8.8 Hz, 1 H) 7.2 (dd, J=7.3, 5.1 Hz, 1 H) 7.3 (d, J=7.6 Hz, 1 H) 7.59 (d, J=8.3 Hz, 2H) 7.7 (m, 1 H) 7.8 (d, J=8.3 Hz, 2 H) 8.0 (d, J=8.8 Hz, 1 H) 8.4 (d, J=4.3 Hz, 1 H) 12.7 (s, 1 H).
4-2 3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-oxazol-2-ylmethyl-amino)-propionic acid: MS MH+ 614; 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.12 (br. s., 2 H) 1.43 (dd, J=6.95, 4.93 Hz, 2 H) 1.59 (d, J=7.33 Hz, 1 H) 1.56 (d, J=3.79 Hz, 2 H) 1.72 (d, J=12.88 Hz, 2 H) 1.69 (br. s., 1 H) 2.01 - 2.12 (m, 3 H) 3.27 (br. s., 2 H) 3.30 (d, J=7.58 Hz, 3 H) 3.85 (s, 3 H) 4.54 (s, 2 H) 6.68 (d, J=8.84 Hz, 1 H) 7.02 (s, 1 H) 7.53 (d, J=8.59 Hz, 2 H) 7.59 - 7.65 (m, 2 H) 7.89 (s, 1 H).
4-3 3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(tetrahydro-furan-2-ylmethyl)-amino]-propionic acid: MS MH+ 617, 1H NMR (400 MHz, DMSO-D6) δ ppm 1.02 - 1.18 (m, 2 H) 1.35 -1.43 (m, 2 H) 1.48 -1.60 (m, 4 H) 1.65 - 1.77 (m, 5 H) 1.80 - 1.86 (m, 1 H) 2.1 -2.19 (m, 3 H) 3.02 - 3.07 (m, 1 H) 3.1-3.33 (m, 4 H) 3.565 (q, J=7.6 Hz, 1 H) 3.675(q, J=7.3 Hz, 1 H) 3.87 (s, 3 H) 3.55 - 3.93 (m,1H) 6.74 (d, J=8.6 Hz, 1H) 7.59 (d, J=8.3 Hz, 2 H) 7.71 (d, J=8.3Hz, 2 H) 7.79 (d, J=8.6 Hz1 1H).
4-4 3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-thiophen-2-ylmethyl-amino)-propionic acid: MS MH+ 629, 1 H NMR (400 MHz, DMSO-D6) δppm 1.12 - 1.17 (m, 2 H) 1.41 - 1.44 (m, 2 H) 1.5 - 1.6 (m, 3 H) 1.65 - 1.80 (m, 3 H) 1.9 - 2.0 (m, 2 H) 2.07 - 2.14 (m, 1 H) 3.24 - 3.29 (m, 2 H) 3.86 (s, 4 H) 4.5 (s, 2 H) 6.72 (d, J=8.6 Hz, 1 H) 6.91 (m, 1 H) 6.95 (m, 1H) 7.37 (m, 1H) 7.59 (d, J=8.6 Hz, 2 H) 7.72 (d, J=8.6 Hz, 2 H) 7.7 (d, J=8.6 Hz, 1H).
4-5 4-[((2-Carboxy-ethyl)-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)- ethyl]-benzenesulfonyl}-amino)-methyl]-benzoic acid: MS MH+ 667, 1H NMR (400 MHz, DMSO- d6) δ ppm 1.12 (dd, J=11.75, 5.94 Hz, 2 H) 1.43 (dd, J=7.07, 4.80 Hz, 2 H) 1.51 - 1.63 (m, 3 H) 1.73 (dd, J=12.88, 6.82 Hz, 3 H) 1.94 (d, J=8.34 Hz, 1 H) 1.91 (br. s., 1 H) 2.12 (ddd, J=13.26, 7.58, 7.45 Hz, 1 H) 3.19 (d, J=19.45 Hz, 2 H) 3.19 (d, J=8.59 Hz, 1 H) 3.87 (s, 4 H) 4.28 (s, 2 H) 6.72 (d, J=8.84 Hz, 1 H) 7.17 (d, J=8.08 Hz, 2 H) 7.60 (d, J=8.34 Hz1 2 H) 7.75 (dd, J=8.21, 5.68 Hz, 5 H).
4-6 3-(Cyclohexylmethyl-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)- ethyl]-benzenesulfonyl}-amino)-propionic acid : MS MH+ 629, 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.76 (br. s., 2 H) 1.08 (br. s., 2 H) 1.11 (d, J=8.59 Hz, 3 H) 1.43 (dd, J=7.07, 4.80 Hz, 3 H) 1.56 (t, J=10.86 Hz, 8 H) 1.66 (br. s., 1 H) 1.79 (t, J=13.89 Hz, 1 H) 1.80 (d, J=13.39 Hz1 1 H) 2.13 (ddd, J=13.26, 7.83, 7.71 Hz, 1 H) 2.32 (d, J=7.83 Hz, 1 H) 2.29 (br. s., 1 H) 2.87 (d, J=7.33 Hz, 2 H) 3.26 (br. s., 1 H) 3.23 (d, J=7.83 Hz, 2 H) 3.90 (s, 3 H) 4.05 (t, J=7.58 Hz, 1 H) 6.87 (d, J=8.59 Hz, 1 H) 7.62 (d, J=8.34 Hz, 2 H) 7.76 (d, J=8.34 Hz, 2 H) 7.98 (d, J=8.84 Hz, 1 H). 4-7 3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(5-methyl-furan-2-ylmethyl)-amino]-propionic acid: MS MH+ 627, 1H NMR (400 MHz, DMSO-d6) δ ppm 1.12 (br. S., 2 H) 1.38-1.49 (m, 2 H) 1.56 (br. s., 3 H) 1.72 (br. s., 3 H) 1.83 (s, 1 H) 2.01 (br. s., 3 H) 2.13 (br. s., 3 H) 3.20 (d, J=6.32 Hz, 2 H) 3.88 (s, 4 H) 4.31 (br. s., 2 H) 5.82 (br. s., 1 H) 6.08 (br. s., 1 H) 6.77 (d, J=8.84 Hz, 1 H) 7.54 - 7.62 (m, 2 H) 7.67 (d, J=8.08 Hz, 2 H) 7.84 (d, J=8.84 Hz, 1 H).
4-8 3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2,5-dimethyl-furan-3-ylmethyl)-amino]-propionic acid: MS MH+ 641, 1H NMR (400 MHz, DMSO-d6) δ ppm 1.11 (br. s., 3 H) 1.40 (br. s., 3 H) 1.69 (br. s., 3 H) 1.88 (br. s., 2 H) 2.11 (s, 8 H) 3.12 (br. s., 3 H) 3.64 (s, 1 H) 3.82 (s, 4 H) 3.99 (s, 2 H) 5.64 (s, 1 H) 6.59 (s, 1 H) 7.56 (s, 3 H) 7.62 (s, 2 H).
4-9 ({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-furan-2-ylmethyl-amino)-acetic acid: MS MH+ 599, 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.07 - 1.18 (m, 1 H) 1.15 (d, J=7.83 Hz, 1 H) 1.44 (dd, J=7.20, 4.67 Hz, 2 H) 1.56 (d, J=7.83 Hz, 3 H) 1.79 (d, J=7.07 Hz, 2 H) 1.74 (d, J=18.69 Hz, 2 H) 2.11 (d, J=13.39 Hz, 1 H) 3.37 (s, 2 H) 3.90 (s, 3 H) 3.98 (br. s., 1 H) 4.57 (s, 2 H) 6.02 (d, J=2.78 Hz, 1 H) 6.19 (dd, J=3.16, 1.89 Hz, 1 H) 6.85 (d, J=8.84 Hz, 1 H) 7.34 (s, 1 H) 7.49 (d, J=8.34 Hz, 2 H) 7.76 (d, J=8.34 Hz, 2 H) 7.95 (d, J=5.81 Hz, 1 H). EC50 in primary enzyme assay 2.4μM
4-10 3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-isopropyl-amino)-propionic acid: MS MH+ 575, 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.93 (d, J=6.82 Hz, 6 H) 1.11 (br. s., 2 H) 1.41 (dd, J=7.07, 4.80 Hz, 2 H) 1.58 (br. s., 2 H) 1.56 (d, J=7.33 Hz, 2 H) 1.68 (br. s., 2 H) 1.75 (dd, J=13.52, 7.20 Hz, 1 H) 2.13 (d, J=12.63 Hz, 1 H) 2.40 (d, J=8.34 Hz, 1 H) 2.37 (br. s., 1 H) 3.23 (d, J=5.31 Hz, 1 H) 3.25 (d, J=7.83 Hz, 1 H) 3.89 (s, 3 H) 3.97 (d, J=7.07 Hz, 1 H) 3.94 (d, J=6.82 Hz, 1 H) 6.83 (d, J=8.84 Hz, 1 H) 7.60 (d, J=8.59 Hz, 2 H) 7.76 (d, J=8.34 Hz, 2 H) 7.92 (d, J=8.59 Hz, 1 H).
4-11 3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 591 , 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.13 (dt, J=11.87, 7.58 Hz, 2 H) 1.42 (dd, J=7.33, 4.80 Hz, 2 H) 1.40 (d, J=1.77 Hz, 1 H) 1.50 - 1.62 (m, 1 H) 1.55 (d, J=5.56 Hz, 2 H) 1.67 - 1.79 (m, 2 H) 2.13 (dd, J=7.45, 5.68 Hz, 1 H) 2.27 (d, J=7.83 Hz, 1 H) 2.24 (br. s., 1 H) 3.14 (s, 3 H) 3.21 - 3.31 (m, 2 H) 3.25 (d, J=5.31 Hz, 3 H) 3.33 - 3.39 (m, 2 H) 3.89 (s, 3 H) 4.00 (t, J=7.45 Hz, 1 H) 6.82 (d, J=8.84 Hz, 1 H) 7.61 (d, J=8.59 Hz, 2 H) 7.75 (d, J=8.34 Hz, 2 H) 7.90 (d, J=8.84 Hz, 1 H).
4-12 3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(1 ,5-dimethyl-1 H-pyrazol-3-ylmethyl)-amino]-propionic acid: MS MH+ 641 , 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.12 (br. s., 2 H) 1.42 (d, J=2.78 Hz, 2 H) 1.72 (br. s., 4 H) 2.05 (s, 4 H) 2.10 (br. s., 3 H) 3.20 (br. s., 3 H) 3.55 (s, 4 H) 3.87 (s, 5 H) 4.18 (s, 2 H) 5.57 (s, 1 H) 6.78 (s, 1 H) 7.57 (s, 2 H) 7.67 (s, 2 H) 7.83 (s, 1 H).
4-13 3-[{4-[2-Cyclopentyl-1 -(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(5-methyl-isoxazol-3-ylmethyl)-amino]-propionic acid: MS MH+ 628, 1H NMR (400 MHz, DMSO-d6) δ ppm 1.12 (br. s., 2 H) 1.41 (br. s., 2 H) 1.70 (br. s., 4 H) 2.06 (s, 4 H) 2.29 (s, 3 H) 3.24 (br. s., 4 H) 3.87 (s, 5 H) 4.35 (s, 2 H) 5.94 (s, 1 H) 6.77 (s, 1 H) 7.60 (s, 2 H) 7.72 (s, 2 H) 7.82 (s, 1 H).
4-14 3-[{4-[2-Cyclopentyl-1 -(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(tetrahydro-pyran-4-ylmethyl)-amino]-propionic acid: MS MH+ 631 , 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.08 (td, J=12.19, 3.66 Hz, 4H) 1.42 (dd, J=7.07, 4.80 Hz, 2H) 1.38 (d, J=8.84 Hz, 1 H) 1.48 - 1.60 (m, 5H) 1.66 - 1.78 (m, 4H) 2.06 (dd, J=10.61 , 5.56 Hz1 2H) 2.06 (br. s., 1 H) 2.91 (d, J=7.33 Hz, 2H) 3.15 - 3.26 (m, 4H) 3.76 - 3.86 (m, 6H) 6.69 (d, J=8.59 Hz, 1 H) 7.58 (d, J=8.34 Hz, 2 H) 7.71 (d, J=8.84 Hz, 1 H) 7.64 - 7.74 (m, 2H).
4-15 3-[{4-[2-Cyclopentyl-1 -(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(4-fluoro-benzyl)-amino]-propionic acid: MS MH+ 641 , 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.12 (br. s., 2 H) 1.43 (br. s., 3 H) 1.56 (br. s., 4 H) 1.70 (d, J=11.37 Hz, 3 H) 2.04 (d, J=8.34 Hz, 2 H) 2.12 (br. s., 1 H) 3.23 (br. s., 2 H) 3.89 (s, 4 H) 4.30 (s, 2 H) 6.82 (d, J=8.34 Hz, 1 H) 7.10 (br. s., 2 H) 7.22 - 7.32 (m, 2 H) 7.62 (d, J=8.34 Hz, 2 H) 7.80 (d, J=7.83 Hz, 2 H) 7.91 (d, 1 H).
4-16 3-({4-[2-Cyclopentyl-1 -(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonylj-cyclopropylmethyl-aminoj-propionic acid: MS MH+ 587, 1 H NMR (400 MHz, DMSO-D6) .δppm 0.11 - 0.18 (m, 2 H) 0.36 - 0.43 (m, 2 H) 0.80 - 0.91 (m, 1 H) 1.05 - 1.16 (m, 2 H) 1.36 - 1.46 (m, 2 H) 1.50 - 1.61 (m, 3 H) 1.65 - 1.77 (m, 3 H) 2.11 (ddd, J=13.26, 7.58, 7.45 Hz, 1 H) 2.17 - 2.25 (m, 2 H) 2.97 (d, J=6.82 Hz, 2 H) 3.28 - 3.39 (m, 2 H) 3.83 - 3.92 (m, 4 H) 6.75 (d, J=8.59 Hz, 1 H) 7.56 - 7.62 (m, 2 H) 7.70 (d, J=8.34 Hz, 2 H) 7.81 (d, J=8.59 Hz, 1 H).
4-17 ({4-[2-Cyclopentyl-1 -(5-methoxy-thiazolo[5,4~b]pyridin-2-ylcarbamoyl)-ethyl]~ benzenesulfonyl}-methyl-amino)-acetic acid: MS MH+ 533, 1 H NMR (400 MHz, DMSO-D6) δ ppm 1.07 - 1.18 (m, 2 H) 1.38 - 1.48 (m, 2 H) 1.51 - 1.63 (m, 3 H) 1.71 (s, 2 H) 1.76 (dd, J=13.39, 6.82 Hz, 2 H) 2.14 (dd, J=7.71 , 5.68 Hz, 1 H) 2.65 (s, 3 H) 3.25 (s, 3 H) 3.90 (s, 3 H) 4.05 (s, 1 H) 6.87 (d, J=8.84 Hz, 1 H) 7.59 (d, J=8.34 Hz, 2 H) 7.75 (d, J=8.34 Hz, 2 H) 7.98 (d, J=8.84 Hz, 1 H).
4-18 3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-methyl-amino)-propionic acid: MS MH+ 547, 1 H NMR (400 MHz, DMSO-D6) δ ppm 1.07 - 1.18 (m, 2 H) 1.37 - 1.48 (m, 2 H) 1.51 - 1.63 (m, 3 H) 1.67 - 1.78 (m, 3 H) 2.10 - 2.19 (m, 1 H) 2.28 (t, J=7.45 Hz, 2 H) 2.65 (s, 3 H) 3.12 (t, J=7.45 Hz, 2 H) 3.89 (s, 3 H) 4.03 (t, J=7.45 Hz, 1 H) 6.84 (d, J=8.84 Hz, 1 H) 7.60 - 7.67 (m, 2 H) 7.69 - 7.75 (m, 2 H) 7.94 (d, J=8.84 Hz, 1 H).
4-19 3-[{4-[2-Cyclopentyl-1 -(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(tetrahydro-pyran-4-yl)-amino]-propionic acid: MS MH+ 617, 1H NMR (400 MHz, DMSO-D6) δ ppm 1.05 - 1.16 (m, 2 H) 1.33 (s, 2 H) 1.36 - 1.46 (m, 2 H) 1.52 - 1.61 (m, 4 H) 1.65 (dd, J=12.38, 4.55 Hz, 2 H) 2.13 (ddd, J=13.07, 7.71 , 7.52 Hz, 1 H) 2.30 - 2.40 (m, 2 H) 3.22 - 3.32 (m, 5 H) 3.73 - 3.85 (m, 3 H) 3.89 (s, 3 H) 4.00 (t, J=7.45 Hz, 1 H) 6.83 (d, J=8.84 Hz, 1 H) 7.60 (d, J=8.34 Hz, 2 H) 7.79 (d, J=8.34 Hz, 2 H) 7.92 (d, J=8.59 Hz, 1 H).
4-20 3-(Cyclohexyl-{4-[(R)-2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)- ethyl]-benzenesulfonyl}-amino)-propionic acid : MS MH+ 615, 1 H NMR (400 MHz, DMSO-d6) δppm 0.97 (s, 2 H) 1.18 (br. s., 3 H) 1.39 (br. s., 5 H) 1.52 (br. s., 4 H) 1.67 (br. s., 5 H) 2.15 (br. s., 3 H) 3.20 (br. s., 4 H) 3.67 (br. s., 2 H) 3.83 (s, 3 H) 6.62 (s, 1 H) 7.56 (s, 3 H) 7.64 (s, 3 H).
4-21 3-[{4-[(R)-2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(4-methoxy-benzyl)-amino]-propionic acid: MS MH+ 653, 1H NMR (400 MHz, DMSO-d6) δ ppm 0.85 (br. s., 2 H) 1.27 (br. s., 2 H) 1.55 (d, J=3.28 Hz, 4 H) 1.84 (br. s., 2 H) 2.11 (br. s., 1 H) 3.14 (br. s., 3 H) 3.70 (s, 4 H) 3.77 (br. s., 1 H) 3.84 (s, 4 H) 4.20 (s, 2 H) 6.67 (s, 1 H) 6.84 (s, 2 H) 7.16 (s, 2 H) 7.60 (s, 2 H) 7.70 (s, 4 H). EC50 in primary enzyme assay 0.92μM
4-22 ({4-[(R)-2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-yIcarbamoyl)-ethyl]- benzenesulfonyl}-methyl-amino)-acetic acid: MS MH+ 533, 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.85 (br. s., 2 H) 1.27 (br. sM 2 H) 1.55 (d, J=3.28 Hz, 4 H) 1.84 (br. s., 2 H) 2.11 (br. s., 1 H) 3.14 (br. s., 3 H) 3.70 (s, 4 H) 3.77 (br. s., 1 H) 3.84 (s, 4 H) 4.20 (s, 2 H) 6.67 (s, 1 H) 6.84 (s, 2 H) 7.16 (s, 2 H) 7.60 (s, 2 H) 7.70 (s, 4 H).
4-23 3-[{4-[(R)-2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-isopropoxy-ethyl)-amino]-propionic acid: MS MH+ 619, 1 H NMR (400 MHz, DMSO-d6) δppm 0.94 (s, 6 H) 1.11 (br. s., 2 H) 1.40 (br. s., 2 H) 1.69 (br. s., 3 H) 2.09 (s, 1 H) 2.22 (br. s., 2 H) 3.21 (s, 6 H) 3.35 (s, 6 H) 3.87 (s, 3 H) 3.94 (br. s., 1 H) 6.79 (s, 1 H) 7.60 (s, 2 H) 7.72 (S, 2 H) 7.84 (s, 1 H).
4-24 3-({4-[(R)-2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-thiazol-2-ylmethyl-amino)-propionic acid: MS MH+ 630, 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.11 (br. s., 2 H) 1.42 (br. s., 2 H) 1.72 (br. s., 3 H) 2.06 (s, 3 H) 3.30 (br. s., 4 H) 3.86 (s, 5 H) 4.66 (s, 2 H) 6.74 (s, 1 H) 7.59 (s, 3 H) 7.65 (s, 1 H) 7.77 (s, 2 H) 7.73 (d, J=8.34 Hz, 3 H).
4-25 3-[{4-[2-Cyclopentyl-1-(5-fluoro-pyridin-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2-methoxy- ethyl)-amino]-propionic acid: MS MH+ 522, 1 H NMR (400 MHz, DMSO-D6) δ ppm 1.08 - 1.18 (m, 2 H) 1.39 - 1.48 (m, 2 H) 1.52 - 1.64 (m, 3 H) 1.70 (dt, J=13.39, 6.69 Hz, 3 H) 2.07 - 2.15 (m, 3 H) 3.14 (s, 3 H) 3.22 - 3.26 (m, 4 H) 3.37 (t, J=5.94 Hz, 3 H) 4.07 (dd, J=8.34, 6.32 Hz, 1 H) 7.61 (d, J=8.34 Hz, 2 H) 7.67 - 7.76 (m, 3 H), 8.1 (dd, J=9.1 , 4.3 Hz, 1 H), 10.9 (s, 1H).
4-26 3-[{4-[(R)-2-Cyclopentyl-1-(pyrimidin-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2-methoxy- ethyl)-amino]-propionic acid: MS MH+ 505, 1 H NMR (400 MHz, DMSO-cf6) δ ppm 1.05-1.19 (m, 2H) 1.38-1.45 (m, 2H) 1.50-1.75 (m, 6H) 2.05-2.14 (m, 1 H) 2.31-2.38 (m, 2H) 3.14 (s, 3H) 3.30- 3.60 (m, 2OH, 6H under H2O) 4.14 (m, 1H) 7.17 (t, J=4.80 Hz, 1 H) 7.61 (d, J=8.34 Hz, 2 H) 7.76 (d, J=8.34Hz, 2 H) 8.63 (d, J=4.80 Hz, 2 H) 10.88 (s, 1 H). 4-27 3-[{4-[(R)-2-Cyclopentyl-1-(4-methyl-thiazol-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-2- methoxy-ethyl)-amino]-propionic acid: MS MH+ 523.
4-28 3-[{4-[(R)-2-Cyclopentyl-1 -(1 -methyl-1 H-pyrazol-3-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid: MS MH+ 507, 1 H NMR (400 MHz, DMSO-cfβj δ ppm 1.04- 1.18 (m, 2H) 1.38-1.47 (m, 2H) 1.50-1.79 (m, 6H) 2.05-2.18 (m, 3H) 3.13 (s, 3H) 3.21-3.39 (m, > 54H (6H under H2O) 3.68 (s, 3H) 3.88-3.90 (m, 1 H) 6.41 (d, J=2.27Hz, 1 H) 7.50 (d, J=2.02Hz, 1 H) 7.57 (d, J=8.59Hz, 2H) 7.72 (d, J = 8.34Hz, 2H).
4-29 3-[{4-[(R)-2-Cyclopentyl-1-(pyrimidin-4-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2-methoxy- ethyl)-amino]-propionic acid: MS MH+ 505, 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.04-1.18 (m, 2H) 1.37-1.48 (m, 2H) 1.49-1.62 (m, 3H) 1.65-1.74 (m, 3H) 2.06-2.17 (m, 1 H) 3.09-3.14 (s, 3H) 3.21-3.40 (m, 13H (6H under H2O) 4.12 (t, J = 7.33Hz, 1 H) 7.60 (d, J = 8.34Hz, 2H) 7.75 (d, J = 8.34Hz, 2H) 8.05 (d, J = 5.56Hz, 1 H) 8.62 (d, J = 5.81 Hz, 1 H) 8.85 (s, 1 H) 11.26 (s, 1 H).
4-30 3-[{4-[(R)-2-Cyclopentyl-1-(4,5-dimethyl-thiazol-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid: MS MH+ 538, 1 H NMR (400 MHz, DMSO-cf6J δ ppm 1.03- 1.17 (m, 2H) 1.37-1.48 (m, 2H) 1.50-1.62 (m, 3H) 1.64-1.76 (m, 3H) 2.07-2.15 (m, 4H) 2.19 (s, 3H) 2.42-2.54 (m, 13H (2H under DMSO) 3.13 (s, 3H) 3.23-3.34 (m, 4H) 3.37 (t, J=5.68Hz) 3.96 (dd, J= 8.46, 6.69Hz, 1 H) 7.58 (d, J=8.34Hz, 2H) 7.78 (d, J=8.34Hz, 2H) 12.18 (s, 1 H).
4-31 3-[{4-[(R)-2-Cyclopentyl-1-(4,5,6,7-tetrahydro-benzothiazol-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 564, 1 H NMR (400 MHz, DMSO-d6) δppm 1.04-1.19 (m, 2H) 1.37-1.48 (m, 2H) 1.50-1.60 (m, 3H) 1.64-1.79 (m, 7H) 2.06- 2.17 (m, 1 H) 2.41-2.48 (m, 2H) 2.42-2.55 (m, 15H, 2H under DMSO) 2.60 (br s, 2H) 3.14 (s, 3H) 3.22-3.48 (m, 6H) 3.98 (dd, J=8.72, 6.44Hz, 1 H) 7.59 (d, J=8.34Hz, 2H) 7.78 (d, J=8.59Hz, 2H) 12.23 (s, 1 H).
4-32 3-[{4-[(R)-2-Cyclopentyl-1-(pyrazin-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2-methoxy- ethyl)-amino]-propionic acid: MS MH+ 505, 1 H NMR (400 MHz, DMSO-c/6) δ ppm C391.05-1.19 (m, 2H) 1.36-1.48 (m, 2H) 1.50-1.65 (m, 3H) 1.67-1.78 (m, 3H) 2.10-2.25 (m, 3H) 3.13 (s, 3H) 3.20-3.29 (m, 4H) 3.37 (t, J=5.94Hz, 2H) 4.12 (t, J=7.45Hz, 1 H) 7.62 (d, J=8.59Hz, 2H) 7.76 (d, J=8.59Hz, 2H) 8.32-8.39 (m, 2H) 9.32 (d, J=1.52Hz, 1 H) 11.14 (s, 1 H). 4-33 3-[{4-[(R)-2-Cyclopentyl-1-(6-fluoro-benzothiazol-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid: MS MH+ 578, 1 H NMR (400 MHz, DMSO-c/6) δ ppm 1.06 - 1.18 (m, 2 H) 1.37 - 1.47 (m, 2 H) 1.51 - 1.63 (m, 3 H) 1.67 - 1.78 (m, 3 H) 2.08 - 2.19 (m, 3 H) 3.15 (s, 3 H) 3.19 - 3.29 (m, 4 H) 3.37 (t, J=5.94 Hz, 2 H) 3.86 - 3.97 (m, 1 H) 7.12 (td, J=9.09, 2.53 Hz, 1 H) 7.52 - 7.63 (m, 3 H) 7.64 - 7.75 (m, 3 H).
4-34 3-[{3-[(R)-1-(5-Chloro-pyrimidin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]-benzenesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid: MS MH+ 539, 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.04-
1.18 (m, 2H) 1.37-1.48 (m, 2H) 1.50-1.63 (m, 3H) 1.65-1.75 (m, 3H) 1.91-2.02 (m, 2H) 2.05-2.14 (m, 1 H) 3.16 (s, 3H) 3.19-3.41 (m, 11 H (6H under H2O) 4.12 (t, J=7.33Hz, 1 H) 7.53 (t, J=7.71 Hz, 1 H) 7.58-7.65 (m, 2H) 7.85 (s, 1 H) 8.72 (s, 2H) 11.48 (br s, 1 H).
4-35 3-[{4-[(R)-1-(5-Chloro-pyrimidin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]-benzenesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid: MS MH+ 539, 1 H NMR (400 MHz, DMSO-c/6) δ ppm 1.04-
1.19 (m, 2H) 1.38-1.47 (m, 2H) 1.50-1.63 (m, 3H) 1.65-1.74 (m, 3H) 2.05-2.14 (m, 1H) 2.25 (t, J=7.71 Hz, 2H) 3.14 (s, 3H) 3.21-3.42 (m, 9H (6H under H2O) 4.12 (t, J=7.35Hz, 1 H) 7.59 (d, J=8.59Hz, 2H) 7.75 (d, J=8.34Hz, 2H) 8.73 (s, 2H) 11.10 (s, 1H).
4-36 3-[{4-[(R)-1-(5-Chloro-thiazol-2-ylcarbamoyl)-2-cyclopentyl-ethyl]-benzenesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid: MS MH+ 544, 1 H NMR (400 MHz, DMSO-D6) δ ppm 1.04 - 1.15 (m, 2 H) 1.36 - 1.46 (m, 2 H) 1.49 - 1.59 (m, 3 H) 1.64 - 1.75 (m, 3 H) 2.09 (ddd, J=13.33, 7.83, 7.64 Hz, 1 H) 2.22 - 2.29 (m, 2 H) 3.15 (s, 3 H) 3.21 - 3.30 (m, 4 H) 3.37 (t, J=5.81 Hz, 2 H) 3.87 (t, J=7.58 Hz, 1 H) 7.30 (s, 1H) 7.57 (d, J=8.59 Hz, 2 H) 7.72 (d, J=8.34 Hz, 2' H).
4-37 3-[{4-[(R)-1-(5-Bromo-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 640, 1H NMR (400 MHz, DMSO-D6) δ ppm 1.04 - 1.16 (m, 2 H) 1.36 - 1.46 (m, 2 H) 1.50 - 1.62 (m, 3 H) 1.65 - 1.74 (m, 3 H) 2.11 (ddd, J=13.14, 7.71 , 7.45 Hz, 1 H) 2.28 - 2.35 (m, 2 H) 3.16 (s, 3 H) 3.21 - 3.30 (m, 4 H) 3.38 (t, J=5.81 Hz, 2 H) 3.72 (t, J=7.58 Hz, 1 H) 7.32 (d, J=8.34 Hz, 1 H) 7.53 - 7.60 (m, 3 H) 7.66 - 7.71 (m, 2 H).
4-38 3-[{4-[2-Cyclopentyl-1-(5-methylsulfanyl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid: : MS MH+ 607, 1H NMR (400 MHz, DMSO-D6) δ ppm 1.04 - 1.15 (m, 2 H) 1.36 - 1.45 (dd, J=7.07, 4.80 Hz, 2 H) 1.51 - 1.63 (m, 2 H) 1.66 - 1.74 (m, 3 H) 2.01 - 2.13 (m, 3 H) 2.5 (s, 3H) 3.17 (s, 3 H) 3.18 - 3.25 (m, 4 H) 3.38 (t, J=6.19 Hz, 2 H) 3.66 (t, J=7.58 Hz, 1 H) 7.01 (d, J=8.3 Hz, 1 H) 7.48 (d, J=8.3 Hz, 1 H) 7.57 (d, J=8.34 Hz, 2 H) 7.65 (d, J=8.3 Hz, 2 H).
4-39 3-[{4-[2-Cyclopentyl-1-(5-methanesulfonyl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 639, 1 H NMR (400 MHz, DMSO-D6) δ ppm 1.05 - 1.17 (m, 2 H) 1.35 - 1.46 (m, 2 H) 1.51 - 1.62 (m, 3 H) 1.66 - 1.77 (m, 3 H) 2.07 - 2.17 (m, 2 H) 2.38 - 2.46 (m, 2 H) 2.74 - 2.82 (m, 1 H) 3.16 (s, 3H) 3.22 (s, 2 H) 3.25 (t, J=5.68 Hz, 2 H) 3.28 - 3.33 (m, 2 H) 3.38 (t, J=5.81 Hz, 2 H) 3.77 (t, J=7.58 Hz, 1 H) 7.60 (d, J=8.34 Hz, 2 H) 7.69 - 7.74 (m, 2 H) 7.75 - 7.82 (m, 2 H).
4-40 3-[{4-[(R)-1-(5-Chloro-quinazoIin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]-benzenesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid: MS MH+ 589, 1 H NMR (400 MHz, DMSO-c/6) δ ppm 1.06- 1.19 (m, 2H) 1.38-1.49 (m, 2H) 1.51-1.60 (m, 3H) 1.63-1.79 (m, 3H) 2.09-2.21 (m, 1 H) 2.29-2.39 (m, 2H) 3.13 (s, 3H) 3.23-3.39 (m, 19H (6H under H2O) 4.27 (s, 1 H) 7.62-7.73 (m, 3H) 7.77 (d, J=8.34Hz, 3H) 7.86-7.94 (m, 1 H) 9.62 (s, 1 H) 11.26 (s, 1 H).
4-41 3-[{4-[2-Cyclopentyl-1-(thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid: MS MH+ 561 , 1 H NMR (400 MHz, DMSO-D6) δ ppm 0.91 - 1.03 (m, 2 H) 1.27 (dd, J=7.20, 4.67 Hz, 2 H) 1.36 - 1.48 (m, 3 H) 1.51 - 1.61 (m, 3 H) 1.97 (dt, J=13.39, 7.58 Hz, 1 H) 2.02 - 2.08 (m, 2 H) 2.98 -3.01 (m, 5 H) 3.05 - 3.14 (m, 4 H) 3.23 (t, J=5.94 Hz, 2 H) 3.65 (t, J=7.45 Hz, 1 H) 7.44 (d, J=8.34 Hz, 2 H) 7.51 - 7.57 (m, 3 H). EC50 in primary enzyme assay 1.2μM
4-42 3-[{4-[(R)-2-Cyclopentyl-1-(5-methylsulfanyl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)- ethyl]-benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 607, 1 H NMR (400 MHz, DMSO-D6) .δppm 1.23 - 1.33 (m, 2 H) 1.53 - 1.64 (m, 2 H) 1.68 - 1.80 (m, 3 H) 1.83 - 1.92 (m, 3 H) 2.24 - 2.35 (m, 3 H) 2.5 (s, 3H) 3.31 - 3.35 (m, 3 H) 3.36 - 3.45 (m, 4 H) 3.51 - 3.58 (m, 2 H) 3.91 (t, J=7.58 Hz, 1 H) 7.25 (d, J=8.34 Hz, 1 H) 7.70 - 7.78 (m, 3 H) 7.82 - 7.87 (m, 2 H). EC50 in primary enzyme assay 0.23μM
4-43 3-[{4-[(R)-1-(5-Chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 595, 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.03 - 1.15 (m, 2 H) 1.35 - 1.46 (m, 2 H) 1.50 - 1.60 (m, 3 H) 1.69 (dt, J=13.14, 6.57 Hz, 3 H) 2.06 - 2.15 (m, 1 H) 2.25 - 2.32 (m, 2 H) 3.15 (s, 3 H) 3.20 - 3.30 (m, 4 H) 3.34 - 3.40 (m, 2 H) 3.76 (s, 1 H) 7.23 (d, J=8.34 Hz, 1 H) 7.59 (d, J=8.34 Hz, 2 H) 7.68 (t, J=8.08 Hz, 3 H).
4-44 3-[(4-{(R)-2-Cyclopentyl-1-[5-(2-methoxy-ethylcarbamoyl)-thiazolo[5,4-b]pyridin-2- ylcarbamoyl]-ethyl}-benzenesulfonyl)-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 662, 1H NMR (400 MHz1 DMSO-c/6) δ ppm 1.10 (td, J=12.00, 8.34 Hz, 2 H) 1.36 - 1.47 (m, 2 H) 1.51 - 1.63 (m, 3 H) 1.66 - 1.75 (m, 3 H) 1.94 - 2.02 (m, 2 H) 2.11 (ddd, J=13.14, 7.58, 7.33 Hz, 1 H) 3.17 (s, 3 H) 3.21 (t, J=6.57 Hz, 4 H) 3.26 - 3.33 (m, 8 H) 3.34 - 3.40 (m, 2 H) 3.42 - 3.49 (m, 4 H) 3.68 (t, J=7.58 Hz, 1 H) 7.61 (td, J=17.31, 8.34 Hz, 5 H) 7.80 - 7.84 (m, 1 H) 8.48 (s, 1H).
4-45 3-[{4-[(R)-2-Cyclopentyl-1-(5-morpholin-4-yl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 646, 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.04-1.19 (m, 2H) 1.37-1.48 (m, 2H) 1.50-1.63 (m, 3H) 1.65-1.79 (m, 3H) 2.06- 2.17 (m, 3H) 3.14 (s, 3H) 3.21-3.50 (m, 28H (10H under H2O) 3.67-3.74 (m, 4H) 3.92-3.97 (m, 1 H) 6.88 (d, J=8.59Hz, 1 H) 7.59 (d, J=8.34Hz, 2H) 7.71-7.79 (m, 3H).
4-46 3-[(4-{(R)-2-Cyclopentyl-1-[5-(2-methoxy-ethoxy)-thiazolo[5,4-b]pyridin-2-ylcarbamoyl]- ethyl}-benzenesulfonyl)-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 635, 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.11 (br. s., 2 H) 1.41 (br. s., 2 H) 1.54 (br. s., 3 H) 1.71 (br. s., 3 H) 2.07 (br. s., 1 H) 2.18 (br. s., 3 H) 3.14 (s, 3 H) 3.22 (br. s., 3 H) 3.28 (s, 4 H) 3.35 (d, J=5.31 Hz, 3 H) 3.62 - 3.72 (m, 2 H) 3.90 (br. s., 1 H) 4.38 (d, J=4.04 Hz, 2 H) 6.77 (d, J=8.59 Hz, 1 H) 7.59 (d, J=8.08 Hz, 2 H) 7.72 (d, J=8.08 Hz, 2 H) 7.83 (br. s., 1 H). EC50 in primary enzyme assay 0.22μM
4-47 3-[{4-[(R)-2-Cyclopentyl-1-(5-ethyl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 589, 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.06-1.17 (m, 2H) 1.24 (t, J=7.58Hz, 3H) 1.37-1.46 (m, 2H) 1.51-1.61 (m, 3H) 1.66-1.79 (m, 3H) 2.08-2.22 (m, 3H) 2.78 (q, J=7.58Hz, 2H) 3.15 (s, 3H) 3.20-3.30 (m, 4H) 3.37 (t, J=5.94Hz, 2H) 3.81-3.90 (m, 1H) 7.15 (d, J=8.34Hz, 1H) 7.59 (d, J=8.34Hz, 2H) 7.71 (d, J=8.59Hz, 3H).
4-48 3-{{4-[(R)-1-(5-Chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-[(R)-1-(tetrahydro-furan-2-yl)methyl]-amino}-propionic acid: MS MH+ 621 , 1 H NMR (400 MHz, DMSO-D6) δ ppm 1.05 - 1.16 (m, 2 H) 1.38 - 1.45 (m, 2 H) 1.48 - 1.60 (m, 5 H) 1.66 - 1.78 (m, 4 H) 1.80 - 1.90 (m, 1 H) 2.08 - 2.15 (m, 2H) 2.37 - 2.46 (m, 2 H) 3.03 (dd, J=14.53, 7.45 Hz, 1 H) 3.18 - 3.29 (m, 2 H) 3.32 - 3.41 (m, 1 H) 3.53 - 3.61 (m, 1 H) 3.65 - 3.72 (m, 1 H) 3.74-3.78 (m, 1H) 3.89 (tt, J=7.11 , 3.51 Hz, 1 H) 7.24 (d, J=8.3 HZ, 1H) 7.59 (d, J=8.59 Hz1 2 H) 7.66 - 7.72 (m, 3 H).
4-49 3-(Benzyl-{4-[(R)-1-(5-chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-amino)-propionic acid: MS MH+ 627 1 H NMR (400 MHz, DMSO-D6) δ ppm 1.06 - 1.17 (m, 2 H) 1.38 - 1.47 (m, 2 H) 1.52 - 1.64 (m, 3 H) 1.72 (ddd, J=13.33, 6.95, 6.76 Hz, 3 H) 2.02 - 2.14 (m, 3 H) 3.19 - 3.27 (m, 2 H) 3.73 (t, J=7.58 Hz, 1 H) 4.30 (s, 2 H) 7.21 - 7.32 (m, 5 H) 7.58 - 7.66 (m, 3 H) 7.74 (d, J=8.59 Hz, 2 H).
4-50 3-[{4-[(R)-2-Cyclopentyl-1-(5-fluoro-thiazo]o[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 579, 1H NMR (400 MHz, DMSO-c/6) δppm 1.09 (d, J=8.08 Hz, 2 H) 1.36 - 1.46 (m, 2 H) 1.53 (br. s., 2 H) 1.60 (s, 1 H) 1.64 - 1.74 (m, 3 H) 2.04 - 2.14 (m, 3 H) 3.11 - 3.18 (m, 3 H) 3.18 - 3.26 (m, 5 H) 3.36 (t, J=6.06 Hz, 3 H) 3.65 (t, J=7.58 Hz, 1 H) 6.85 (dd, J=8.59, 1.77 Hz, 1 H) 7.53 - 7.58 (m, 2 H) 7.62 - 7.73 (m, 3 H).
4-51 3-[{4-[(S)-1-(5-Chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 595, 1 H NMR (400 MHz, DMSO-c/6) δ ppm 1.10 (br. s., 2 H) 1.41 (br. s., 2 H) 1.54 (br. s., 2 H) 1.69 (br. s., 2 H) 2.08 (d, J=5.05 Hz, 1 H) 2.24 (br. s., 1 H) 3.15 (s, 3 H) 3.19 - 3.31 (m, 7 H) 3.37 (t, J=5.94 Hz, 5 H) 3.67 (s, 1 H) 7.17 (d, J=8.34 Hz, 1 H) 7.58 (t, J=9.22 Hz, 3 H) 7.67 (d, J=8.34 Hz, 2 H). EC50 in primary enzyme assay 44μM
4-52 3-{{4-[(R)-1-(5-Chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclohexyl-ethyl]- benzenesulfonyl}-[(R)-1-(tetrahydro-furan-2-yl)methyl]-amino}-propionic acid: MS MH+ 635, 1 H NMR (400 MHz, DMSO-c/6) δ ppm 0.90 (br. s., 2 H) 1.09 (br. s., 4 H) 1.52 - 1.63 (m, 5 H) 1.74 (br. s., 4 H) 1.83 (d, J=11.62 Hz, 1 H) 1.94 - 2.05 (m, 1 H) 2.32 (br. s., 2 H) 3.03 (dd, J=14.40, 7.33 Hz, 1 H) 3.19 - 3.28 (m, 3 H) 3.34 (d, J=7.58 Hz, 2 H) 3.52 - 3.61 (m, 1 H) 3.62 - 3.72 (m, 1 H) 3.74 - 3.83 (m, 1 H) 3.84 - 3.93 (m, 1 H) 7.19 (d, J=8.34 Hz1 1 H) 7.54 - 7.64 (m, 3 H) 7.65 - 7.70 (m, 2 H). 4-53 3-(Benzyl-{4-[(R)-1-(5-chloro-thiazolo[5,4-b]pyridin-2-yIcarbamoyl)-2-cyclohexyl-ethyl]- benzenesulfonyl}-amino)-propionic acid: MS MH+ 641 , 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.91 (br. s., 2 H) 1.10 (d, J=5.31 Hz, 4 H) 1.51 - 1.59 (m, 2 H) 1.62 (d, J=12.38 Hz, 2 H) 1.74 (br. s., 2 H) 1.86 - 1.96 (m, 2 H) 1.96 - 2.06 (m, 1 H) 3.15 - 3.26 (m, 2 H) 3.78 (t, J=7.58 Hz, 1 H) 4.28 (s, 2 H) 7.17 (d, J=8.34 Hz, 1 H) 7.21 - 7.32 (m, 5 H) 7.59 (dd, J=8.34, 6.06 Hz, 3 H) 7.71 (d, J=8.34 Hz, 2 H).
4-54 2-[(R)-2-(4-{(2-Carboxy-ethyl)-[(R)-1-(tetrahydro-furan-2-yl)methyl]-sulfamoyl}-phenyl)-3- cyclopentyl-propionylamino]-benzothiazole-6-carboxylic acid ethyl ester: MS MH+ 658, 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.09 (br. s., 2 H) 1.32 (t, J=7.07 Hz, 3 H) 1.40 (br. s., 2 H) 1.53 (br. s., 4 H) 1.70 (br. s., 5 H) 1.81 (br. s., 1 H) 2.12 (br. s., 3 H) 2.98 - 3.09 (m, 1 H) 3.10 - 3.20 (m, 3 H) 3.23 (br. s., 1 H) 3.28 (br. s., 2 H) 3.57 (d, J=7.33 Hz1 2 H) 3.64 - 3.74 (m, 2 H) 3.91 (br. s., 1 H) 4.27 (q, J=7.07 Hz, 2 H) 7.39 (d, J=8.34 Hz, 1 H) 7.57 (d, J=8.08 Hz, 2 H) 7.61 - 7.69 (m, 2 H) 7.71 - 7.79 (m, 1 H) 8.19 (br. s., 1 H).
4-55 2-[(R)-2-(4-{(2-Carboxy-ethyl)-[(R)-1-(tetrahydro-furan-2-yl)methyl]-sulfamoyl}-phenyl)-3- cyclopentyl-propionylamino]-benzothiazole-6-carboxylic acid: MS MH+ 630, 1 H NMR (400 MHz, DMSO-d6) δppm 1.05 - 1.16 (m, 2 H) 1.41 (dd, J=7.07, 4.80 Hz, 4 H) 1.50 - 1.58 (m, 2 H) 1.67 - 1.75 (m, 3 H) 1.80 - 1.86 (m, 3 H) 1.93 - 2.02 (m, 2 H) 2.05 - 2.15 (m, 1 H) 2.99 - 3.06 (m, 1 H) 3.12 - 3.20 (m, 2 H) 3.22 - 3.31 (m, 2 H) 3.55 - 3.63 (m, 2 H) 3.65 - 3.73 (m, 1 H) 3.88 - 3.95 (m,
1 H) 7.14 (d, J=8.08 Hz, 1 H) 7.54 - 7.58 (m, 2 H) 7.60 - 7.66 (m, 2 H) 7.99 (d, J=1.26 Hz, 1 H).
4-56 3-[{4-[(R)-1-(5-Chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclohexyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyI)-amino]-propionic acid: MS MH+ 609, 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.90 (br. s., 2 H) 1.09 (br. s., 4 H) 1.58 (dd, J=13.89, 6.57 Hz, 3 H) 1.72 (br. s., 3 H) 1.95 - 2.06 (m, 1 H) 2.26 - 2.36 (m, 2H) 3.15 (s, 3 H) 3.20 - 3.31 (m, 4 H) 3.37 (t, J=5.56 Hz,
2 H) 3.88 (t, J=7.20 Hz, 1 H) 7.25 (d, J=8.34 Hz, 1 H) 7.58 (d, J=8.34 Hz, 2 H) 7.70 (d, J=8.34 Hz, 3 H).
4-57 3-[{4-[(R)-2-Cyclopentyl-1-(5-pyridin-4-yl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 638, 1 H NMR (400 MHz, DMSO-cy6) δ ppm 1.10-1.19 (m, 2H) 1.37-1.48 (m, 2H) 1.51-1.64 (m, 3H) 1.67-1.79 (m, 3H) 2.08- 2.17 (m, 1 H) 2.32-2.45 (m, 2H) 3.16 (s, 3H) 3.22-3.46 (m, 21H (6H under H2O) 3.84 (br s, 1 H) 7.62 (d, J=8.34Hz, 2H) 7.73 (d, J=8.34Hz, 2H) 7.80-7.90 (br s, 1 H) 7.98-8.08 (m, 3H) 8.63 (d, J=5.81 Hz, 2H).
4-58 3-[{4-[(R)-1-(5-Chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid tert-butyl ester: MS MH+ 651 , 1 H NMR (400 MHz1 ACETONITRILE-d3) δppm 1.09 - 1.18 (m, 4 H) 1.35 (s, 9 H) 1.40 - 1.51 (m, 2 H) 1.55 - 1.66 (m, 3 H) 1.68 - 1.77 (m, 2 H) 1.91 (dt, J=4.86, 2.49 Hz, 2 H) 2.08 - 2.10 (m, 3 H) 2.13 - 2.21 (m, 1 H) 2.39 - 2.45 (m, 2 H) 3.16 (s, 3 H) 3.22 - 3.29 (m, 2 H) 3.32 - 3.40 (m, 4 H) 3.95 (t, 1 H) 7.41 (d, J=8.59 Hz, 1 H) 7.57 - 7.63 (m, 2 H) 7.73 - 7.79 (m, 2 H) 7.95 (d, J=8.59 Hz, 1 H) 10.30 (br. s., 1 H).
4-59 3-[{4-[(R)-1-(5-Chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid benzyl ester: MS MH+ 685, 1H NMR (400 MHz, DMSO-d6). ppm 1.10 (d, J=3.54 Hz, 2 H) 1.15 (dd, J=11.49, 6.95 Hz1 2 H) 1.42 (dd, J=7.07, 4.80 Hz, 1 H) 1.50 - 1.62 (m, 2 H) 1.64 - 1.74 (m, 2 H) 1.75 - 1.85 (m, 1 H) 2.15 (dd, J=7.58, 5.81 Hz, 1 H) 2.63 (t, J=7.33 Hz, 2 H) 3.11 (s, 3 H) 3.24 - 3.32 (m, 2 H) 3.33 - 3.42 (m, 4 H) 4.11 (t, J=7.58 Hz, 1 H) 5.04 (s, 2 H) 7.28 - 7.37 (m, 5 H) 7.55 (d, J=8.34 Hz, 1 H) 7.63 (d, J=8.59 Hz, 2 H) 7.81 (d, J=8.34 Hz, 2 H) 8.14 (d, J=8.59 Hz, 1 H) 12.97 (s, 1 H).
4-60 3-[(4-{(S)-2-Cyclopentyl-1-[5-(2-methoxy-ethylcarbamoyl)-thiazolo[5,4-b]pyridin-2- ylcarbamoyl]-ethyl}-benzenesulfonyl)-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ 662, 1 H NMR (400 MHz, DMSO-c/6) δ ppm 1.11 (br. s., 2 H) 1.35 - 1.47 (m, 2 H) 1.51 - 1.63 (m, 3 H) 1.65 - 1.76 (m, 3 H) 2.12 (dd, J=14.15, 6.57 Hz, 2 H) 3.16 <s, 3 H) 3.20 - 3.29 (m, 8 H) 3.37 (t, J=6.06 Hz, 4 H) 3.41 - 3.50 (m, 4 H) 3.70 (t, J=7.58 Hz, 1 H) 7.58 (d, J=8.59 Hz, 2 H) 7.61 - 7.69 (m, 3 H) 7.83 (d, J=8.34 Hz, 1 H) 8.48 (s, 1 H). EC50 in primary enzyme assay 6.0μM
4-61 3-(Carboxymethyl-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)- ethyl]-benzenesulfonyl}-amino)-propionic acid: MS MH+ 591, 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.10 (br. s., 2 H) 1.39 (br. s., 3 H) 1.69 (br. s., 4 H) 2.07 (s, 1 H) 2.20 (br. s., 2 H) 3.25 (br. s., 4 H) 3.40 (s, 3 H) 3.65 (br. s., 1 H) 3.82 (s, 4 H) 6.61 (s, 1 H) 7.51 - 7.61 (m, 3 H) 7.65 (s, 2 H). 4-62 2-{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonylamino}-2-methyl-propionic acid: MS MH+ 547, 1H NMR (400 MHz, DMSO-D6) δ ppm 1.06 - 1.16 (m, 8 H) 1.37 - 1.46 (m, 2 H) 1.56 (ddd, J=14.53, 7.20, 7.07 Hz, 3 H) 1.64 - 1.71 (m, 2 H) 1.71 - 1.81 (m, 1 H) 2.11 (ddd, J=13.20, 7.83, 7.52 Hz, 1 H) 3.90 (s, 3 H) 4.01 (t, J=7.33 Hz, 1 H) 6.87 (d, J=8.84 Hz, 1 H) 7.54 (d, J=8.34 Hz, 2 H) 7.74 (d, J=8.34 Hz, 2 H) 7.99 (d, J=8.84 Hz, 1 H).
4-63 Succinic acid mono-[2-({4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2- ylcarbamoyl)-ethyl]-benzenesulfonyl}-methyl-amino)-ethyl] ester: MS MH+ 619, 1 H NMR (400 MHz, DMSO-D6) δ ppm 1.06 - 1.17 (m, 2 H) 1.37 - 1.48 (m, 2 H) 1.51 - 1.62 (m, 3 H) 1.72 (ddd, J=13.33, 6.95, 6.76 Hz, 3 H) 2.07 - 2.18 (m, 1 H) 2.19 - 2.29 (m, 2 H) 2.29 - 2.37 (m, 2 H) 2.74 (s, 3 H) 3.20 - 3.31 (m, 2 H) 3.84 - 3.91 (m, 3 H) 3.99 (t, J=7.33 Hz, 1 H) 4.06 (t, J=5.43 Hz, 2 H) 6.79 (d, J=8.84 Hz, 1 H) 7.60 (d, J=8.34 Hz, 2 H) 7.73 (d, J=8.59 Hz, 2 H) 7.87 (d, J=8.84 Hz, 1 H).
4-64 (S)-2-{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonylamino}-propionic acid: MS MH+ 533, 1H NMR (400 MHz, DMSO-D6) δ ppm 1.11 (s, 2 H) 1.15 (dd, J=6.82, 2.53 Hz, 4 H) 1.43 (dd, J=6.06, 4.55 Hz, 2 H) 1.56 (d, J=6.32 Hz1 3 H) 1.65 - 1.77 (m, 3 H) 2.07 - 2.16 (m, 1 H) 2.89 - 2.96 (m, 1 H) 3.86 - 3.95 (m, 4 H) 6.80 (d, J=8.84 Hz, 1 H) 7.52 - 7.60 (m, 2 H) 7.70 (d, J=7.83 Hz, 2 H) 7.88 (s, 1 H).
4-65 3-((2-Carboxy-ethyl)-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)- ethyl]-benzenesulfonyl}-amino)-propionic acid: MS MH+ 605, 1 H NMR (400 MHz, DMSO-d6) δppm 1.09 (br. s., 3 H) 1.41 (br. s., 3 H) 1.53 (br. s., 3 H) 2.09 (br. s., 6 H) 3.20 (br. s., 5 H) 3.74 (s, 1 H) 3.83 (s, 4 H) 6.65 (s, 1 H) 7.57 (s, 2 H) 7.60 - 7.68 (m, 4 H).
4-66 {4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonylamino}-acetic acid: MS MH+ 519, 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.11 (br. s., 3 H) 1.41 (br. s., 3 H) 1.70 (br. s., 4 H) 2.08 (s, 1 H) 2.94 (s, 2 H) 3.86 (s, 5 H) 6.75 (s, 1 H) 7.55 (s, 2 H) 7.69 (s, 3 H) 7.80 (s, 1 H).
4-67 4-{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonylaminoj-butyric acid: MS MH+ 547, 1H NMR (400 MHz, DMSO-D6) δ ppm 1.10 - 1.16 (m, 2 H) 1.35 - 1.43 (m, 2 H) 1.46 - 1.61 (m, 5H) 1.65 - 1.76 (m, 3 H) 2.0 (t, J=6.7 Hz, 2 H) 2.1 (ddd, J=13.1 , 7.7, 7.5 Hz, 1 H) 2.7 (t, J=6.6 Hz, 2 H) 3.8 (t, J=7.5 Hz, 1 H) 3.9 (s, 3 H) 6.7 (d, J=8.6 Hz, 1 H) 7.55 (d, J=8.3 Hz, 2 H) 7.68(d, J=8.3 Hz, 2 H) 7.74 (d, J=8.8 Hz, 1 H).
4-68 (Carboxymethyl-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-amino)-acetic acid: MS MH+ 577, 1 H NMR (400 MHz, DMSO-d6) δ ppm 1.07 (br. s., 3 H) 1.41 (br. s., 3 H) 1.70 (br. s., 4 H) 2.06 (s, 1 H) 3.59 (s, 5 H) 3.82 (s, 4 H) 6.55 (s, 1 H) 7.40 (s, 2 H) 7.53 (s, 1 H) 7.74 (s, 2 H).
4-69 3-[{4-[2-Cyclopentyl-1-(5-trifluoromethyl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid: MS MH+ = 629, 1 H NMR (400 MHz, DMSO-D6) δ ppm 0.98 - 1.09 (m, 2 H) 1.29 - 1.40 (m, 2 H) 1.44 - 1.56 (m, 3 H) 1.58 - 1.68 (m, 3 H) 1.91 - 1.97 (m, 2 H) 2.05 (dt, J=13.20, 7.42 Hz, 1 H) 3.10 (s, 3 H) 3.12 - 3.18 (m, 4 H) 3.31 (t, J=6.19 Hz, 3 H) 3.63 (t, J=7.58 Hz, 1 H) 7.47 - 7.53 (m, 3 H) 7.57 - 7.61 (m, 3 H);
4-70 3-[{2-Chloro-4-[1-(5-chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid MS MH+ = 629 1 H NMR (400 MHz, DMSO-CZ6) δ ppm 1.11 (br. s., 2 H) 1.41 (d, J=2.27 Hz, 2 H) 1.50 - 1.62 (m, 3 H) 1.64 - 1.74 (m, 1 H) 1.69 (d, J=6.06 Hz, 2 H) 2.08 (dd, J=7.45, 5.94 Hz, 1 H) 2.26 (br. s., 2 H) 3.13 (s, 3 H) 3.40 (br. s., 1 H) 3.38 (d, J=5.05 Hz, 3 H) 3.44 (d, J=15.41 Hz, 3 H) 3.67 (t, J=7.71 Hz, 1 H) 7.18 (d, J=8.34 Hz, 1 H) 7.48 (dd, J=8.34, 1.52 Hz, 1 H) 7.64 (d, J=1.52 Hz, 1 H) 7.61 (d, J=8.34 Hz, 1 H) 7.85 (d, J=8.08 Hz, 1 H)
4-71 3-[{4-[2-Cyclopentyl-1-(5-ethyl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]-2- trifluoromethyl-benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid MS MH+ = 657 1 H NMR (400 MHz, DMSOd6) δ ppm 1.12 (ddd, J=16.74, 11.68, 8.21 Hz, 2 H) 1.24 (t, J=7.58 Hz, 3 H) 1.42 (dd, J=7.20, 4.67 Hz, 2 H) 1.50 - 1.61 (m, 3 H) 1.69 (d, J=5.05 Hz, 2 H) 2.15 (ddd, J=13.33, 7.71 , 7.52 Hz, 1 H) 2.32 - 2.39 (m, 1 H) 2.35 (d, J=7.58 Hz, 1 H) 2.79 (q, J=7.58 Hz, 2 H) 3.16 (s, 3 H) 3.39 - 3.50 (m, 2 H) 3.43 (dd, J=12.76, 4.42 Hz, 5 H) 3.99 (t, J=7.58 Hz, 1 H) 7.18 (d, J=8.08 Hz, 1 H) 7.75 (d, J=8.34 Hz, 1 H) 7.85 (dd, J=8.34, 1.26 Hz, 1 H) 7.97 (d, J=I .26 Hz, 1 H) 7.92 - 7.98 (m, 1 H)

Claims

What is claimed is:
1. A compound of the formula
Figure imgf000062_0001
wherein
Q combined together with the carbon and nitrogen atoms to which it is attached form a 5- to 6-membered monocyclic heteroaromatic ring; or
Q combined together with the carbon and nitrogen atoms to which it is attached form a 9- to 10-membered bicyclic heterocycle;
R1 and R2 are, independently from each other, hydrogen, halogen, cyano, nitro, optionally substituted alkyl, alkoxy, alkylthio, alkylthiono, sulfonyl, free or esterified carboxy, carbamoyl, sulfamoyl, optionally substituted amino, aryl or heterocyclyl; or
R2 is absent;
R3 is (C3-6)cycloalkyl or (C3.6)heterocyclyl;
R4 is hydrogen, halogen, cyano, lower alkyl or lower alkoxy;
R5 is (C3-12)cycloalkyl, (C6-10)aryl, (C3-10)heterocyclyl or (C1-6)alkyl optionally substituted by (Ci-6)alkoxy, (C1-2)alkoxy-(C1-4)alkoxy, (C1-6)alky!thio, (C3-7)cycloalkyl, (C3-7)cycloalkoxy, (C3.7)cycloalkylthio, (C6-io)aryl, (C6-io)aryloxy, (C6-io)arylthio, (C3-io)heterocyclyl or (C3-10)heterocyclyloxy;
R6 is free or esterified carboxy, tetrazolyl, cyano or -C(O)NR7R8 in which
R7 and Re are, independently from each other, hydrogen, optionally substituted alkyl, cycloalkyl, aryl or heterocyclyl; or
R7 and Rs combined are alkylene which together with the nitrogen atom to which they are attached form a 4- to 7-membered ring; n is an integer from 1 to 6; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
2. A compound according to Claim 1 , wherein R3 is cyclopentyl;
R4 is hydrogen;
R6 is free or esterified carboxy; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
3. A compound according to Claim 2 of the formula
Figure imgf000063_0001
wherein
R9 hydrogen, optionally substituted lower alkyl, lower alkenyl, cycloalkyl, aryl or aralkyl;
Q combined together with the carbon and nitrogen atoms to which it is attached form a 5- to 6-membered monocyclic heteroaromatic ring which is selected from the group consisting of
Figure imgf000063_0002
or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
4. A compound according to Claim 3, wherein n is an integer from 1 to 3; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
5. A compound according to Claim 4, wherein R9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
(
6. A compound according to Claim 4, wherein
R1 is hydrogen, halogen, cyano, trifluoromethyl, alkoxy, alkylthio or carboxy;
R2 is absent; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
7. A compound according to Claim 6, wherein
Q combined together with the carbon and nitrogen atoms to which it is attached form a 5- to 6-membered monocyclic heteroaromatic ring which is selected from the group consisting of
Figure imgf000064_0001
or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
8. A compound according to Claim 7, wherein
R5 is (C3-7)cycloalkyl or (C1-4)alkyl substituted by (C1-6)alkoxy, (C3-7)cycloalkyl, (C6-10)aryl or (C5-6)heterocyclyl; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
9. A compound according to Claim 8, wherein R9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
10. A compound according to Claim 9, wherein (C1-4)alkyl is methyl or ethyl; (Ci-6)alkoxy is methoxy or ethoxy; (C6-io)aryl is optionally substituted phenyl; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
11. A compound according to Claim 2 of the formula
Figure imgf000065_0001
wherein
R9 is hydrogen, optionally substituted lower alkyl, lower alkenyl, cycloalkyl, aryl or aralkyl;
Q combined together with the carbon and nitrogen atoms to which it is attached form a 9- to 10-membered bicyclic heterocycle which is selected from the group consisting of
Figure imgf000065_0002
or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
12. A compound according to Claim 11 , wherein n is an integer from 1 to 3; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
13. A compound according to Claim 12, wherein R9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
14. A compound according to Claim 12, wherein
Ri is hydrogen, halogen, cyano, trifluoromethyl, alkoxy, alkylthio or carboxy; R2 is absent; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
15. A compound according to Claim 14, wherein
Q combined together with the carbon and nitrogen atoms to which it is attached form a 9- to 10-membered bicyclic heterocycle which is selected from the group consisting of
Figure imgf000066_0001
or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
16. A compound according to Claim 15, wherein
R5 is (C3-7)cycloalkyl or (C1-4)alkyl substituted by (C-|.6)alkoxy, (C3-7)cycloalkyl, (C6-10)aryl or (C5-6)heterocyclyl; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
17. A compound according to Claim 16, wherein R9 is hydrogen; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
18. A compound according to Claim 17, wherein (C1-4)alkyl is methyl or ethyl;
(Ci-6)alkoxy is methoxy or ethoxy; (C6-io)aryl is optionally substituted phenyl; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
19. A method for the activation of glucokinase activity in mammals which method comprises administering to a mammal, in need thereof, a therapeutically effective amount of a compound of Claim 1.
20. A method for the treatment of conditions associated with glucokinase activity in mammals which method comprises administering to a mammal, in need thereof, a therapeutically effective amount of a compound of Claim 1.
21. A method according to Claim 20, which method comprises administering a therapeutically effective amount of said compound in combination with a therapeutically effective amount of an anti-diabetic agent, a hypolipidemic agent, an anti-obesity agent or an anti-hypertensive agent.
22. A method for the treatment of impaired glucose tolerance, type 2 diabetes and obesity which method comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Claim 1.
23. A pharmaceutical composition comprising a therapeutically effective amount of a compound of Claim 1 in combination with one or more pharmaceutically acceptable carriers.
24. A pharmaceutical composition according to Claim 23 for the treatment of impaired glucose tolerance, type 2 diabetes and obesity.
25. A pharmaceutical composition comprising a therapeutically effective amount of a compound of Claim 1 in combination with a therapeutically effective amount of an antidiabetic agents, a hypolipidemic agent, an anti-obesity agent or an anti-hypertensive agent.
26. A pharmaceutical composition according to Claim 25 for the treatment of impaired glucose tolerance, type 2 diabetes and obesity.
27. A pharmaceutical composition according to Claim 23 or 25, for use as medicament.
28. Use of a pharmaceutical composition according to Claim 23 or 25, for the preparation of a medicament for the treatment of conditions associated with glucokinase activity.
29. Use of a compound according to Claim 1 , for the preparation of a pharmaceutical composition for the treatment of conditions associated with glucokinase activity.
30. Use according to claim 28 or 29, wherein the condition associated with glucokinase activity is selected from impaired glucose tolerance, type 2 diabetes and obesity.
31. A compound according to Claim 1 , for use as a medicament.
32. A compound of the formula
Figure imgf000068_0001
wherein
Q combined together with the carbon and nitrogen atoms to which it is attached form a 5- to 6-membered monocyclic heteroaromatic ring; or
Q combined together with the carbon and nitrogen atoms to which it is attached form a 9- to 10-membered bicyclic heterocycle;
R1 and R2 are, independently from each other, hydrogen, halogen, cyano, nitro, optionally substituted alkyl, optionally substituted alkenyl, alkynyl, alkoxy, alkylthio, alkylthiono, sulfonyl, free or esterified carboxy, carbamoyl, sulfamoyl, optionally substituted amino, aryl, aryloxy or heterocyclyl;
R3 is (C3-6)cycloalkyl or (C3.6)heterocyclyl;
R4 is hydrogen, halogen, cyano, lower alkyl or lower alkoxy;
R5 is hydrogen, (C3.12)cycloalkyl, (C6-10)aryl, (C3-10)heterocyclyl or (C1-6)alkyl optionally substituted by carboxy, (C1^aIkOXy, (C1-2)alkoxy-{C1-4)alkoxy, (C1-6)alkylthio, (C3- 7)cycloalkyl, (C3-7)cycloalkoxy, (C3-7)cycloalkylthio, (C6-1o)aryl, (C6-10)aryloxy, (C6. io)arylthio, (C3-i0)heterocyclyl or (Ca-^heterocyclyloxy;
R6 is free or esterified carboxy, tetrazolyl, cyano or -C(O)NR7R8 in which
R7 and R8 are, independently from each other, hydrogen, optionally substituted alkyl, cycloalkyl, aryl or heterocyclyl; or
R7 and R8 combined are alkylene which together with the nitrogen atom to which they are attached form a 4- to 7-membered ring; and n is an integer from 1 to 6; or an enantiomer thereof; or an enantiomeric mixture thereof; or a pharmaceutically acceptable salt thereof.
33. The compound according to claim 32, wherein the compound is selected from the group consisting of:
3-(Benzyl-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-amino)-propionic acid;
3-[{4-[2-Cyclopentyl-1-([1 ,3,4]thiadiazol-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid;
3-(Benzofuran-2-ylmethyl-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2- ylcarbamoyl)-ethyl]-benzenesulfonyl}-amino)-propionic acid;
3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-pyridin-2-ylmethyl-amino)-propionic acid;
3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-oxazol-2-ylrnethyl-amino)~propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(tetrahydro-furan-2-ylmethyl)-amino]-propionic acid;
3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyI}-thiophen-2-ylmethyl-amino)-propionic acid;
4-[((2-Carboxy-ethyl)-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2- ylcarbamoyO-ethy^-benzenesulfonylJ-aminoJ-methylJ-benzoic acid;
3-(Cyclohexylmethyl-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2- ylcarbamoyl)-ethyl]-benzenesulfonyl}-amino)-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(5-methyl-furan-2-ylmethyl)-amino]-propionic acid; 3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyI]- benzenesulfonyl}-(2,5-dimethyl-furan-3-ylmethyl)-amino]-propionic acid;
({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-furan-2-ylmethyl-amino)-acetic acid;
3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-isopropyl-amino)-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(1 ,5-dimethyl-1 H-pyrazol-3-ylmethyl)-amino]-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(5-methyl-isoxazol-3-ylmethyl)-amino]-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(tetrahydro-pyran-4-ylmethyl)-annino]-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyI}-(4-fluoro-benzyl)-amino]-propionic acid;
3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-cyclopropylmethyl-amino)-propionic acid;
({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-methyl-amino)-acetic acid;
3-({4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-methyl-amino)-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyI]- benzenesulfonyl}-(tetrahydro-pyran-4-yl)-amino]-propionic acid;
3-(Cyclohexyl-{4-[(R)-2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2- ylcarbamoyO-ethylJ-benzenesulfonylJ-aminoJ-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(4-methoxy-benzyl)-amino]-propionic acid;
({4-[(R)-2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonylj-methyl-aminoj-acetic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyI]- benzenesulfonyl}-(2-isopropoxy-ethyl)-amino]-propionic acid;
3-({4-[(R)-2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonylHhiazol^-ylmethyl-amino^propionic acid; 3-[{4-[2-Cyclopentyl-1-(5-fluoro-pyridin-2-ylcarbamoyl)-ethyl]-ben2enesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(pyrimidin-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid;
3-[{4~[(R)-2-Cyclopentyl-1-(1-methyl-1H-pyrazol-3-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(pyrimidin-4-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(4,5-dimethyl-thiazol-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(4,5,6,7-tetrahydro-benzothiazol-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(pyrazin-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}-(2- methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(6-fluoro-benzothiazoI-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{3-[(R)-1-(5-Chioro-pyrimidin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]-benzenesulfonyl}- (2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-1-(5-Chloro-pyrimidin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]-benzenesulfonyl}- (2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-1-(5-Chloro-thiazol-2-ylcarbamoyl)-2-cyclopentyl-ethyl]-benzenesulfonyl}- (2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-1-(5-Bromo-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methylsulfanyl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-methanesulfonyl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)- ethyl]-benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-1-(5-Chloro-quinazolin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[2-Cyclopentyl-1-(thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]-benzenesulfonyl}- (2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(5-methylsulfanyl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)- ethyl]-benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid; 3-[{4-[(R)-1-(5-Chloro-thiazolo[5, 4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[(4-{(R)-2-Cyclopentyl-1-[5-(2-methoxy-ethylcarbamoyl)-thiazolo[5,4-b]pyridin-2- ylcarbamoyl]-ethyl}-benzenesulfonyl)-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(5-morpholin-4-yl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)- ethyl]-benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[(4-{(R)-2-Cyclopentyl-1-[5-(2-methoxy-ethoxy)-thiazolo[5,4-b]pyridin-2- ylcarbamoyl]-ethyl}-benzenesulfonyl)-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(5-ethyl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-{{4-[(R)-1-(5-Chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-[(R)-1-(tetrahydro-furan-2-yl)methyl]-annino}-propionic acicl;
3-(Benzyl-{4-[(R)-1-(5-chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyI- ethyl]-benzenesulfonyl}-amino)-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(5-fluoro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(S)-1-(5-Chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-{{4-[(R)-1-(5-Chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclohexyl-ethyl]- benzenesulfonyl}-[(R)-1-(tetrahydro-furan-2-yl)methyl]-amino}-propionic acid;
3-(Benzyl-{4-[(R)-1-(5-chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclohexyl- ethyl]-benzenesulfonyl}-amino)-propionic acid;
2-[(R)-2-(4-{(2-Carboxy-ethyl)-[(R)-1-(tetrahydro-furan-2-yl)methyl]-sulfamoyl}- phenyl)-3-cyclopentyl-propionylamino]-benzothiazole-6-carboxylic acid ethyl ester;
2-[(R)-2-(4-{(2-Carboxy-ethyl)-[(R)-1-(tetrahydro-furan-2-yl)methyl]-sulfamoyl}- phenyl)-3-cyclopentyl-propionylamino]-benzothiazole-6-carboxylic acid;
3-[{4-[(R)-1-(5-Chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclohexyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-2-Cyclopentyl-1-(5-pyridin-4-yl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[(R)-1-(5-Chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid tert-butyl ester;
3-[{4-[(R)-1-(5-Chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid benzyl ester; 3-[(4-{(S)-2-Cyclopentyl-1-[5-(2-methoxy-ethylcarbamoyl)-thiazolo[5,4-b]pyridin-2- ylcarbamoyl]-ethyl}-benzenesulfonyl)-(2-methoxy-ethyl)-amino]-propionic acid;
3-(Carboxymethyl-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2- ylcarbamoyl)-ethyl]-benzenesulfonyl}-amino)-propionic acid;
2-{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonylamino}-2-methyl-propionic acid;
Succinic acid mono-[2-({4-[2-cyclopentyl-1 -(5-methoxy-thiazolo[5,4-b]pyridin-2- ylcarbamoyl)-ethyl]-benzenesulfonyl}-methyl-amino)-ethyl] ester;
(S)-2-{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonylamino}-propionic acid;
3-((2-Carboxy-ethyl)-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2- ylcarbamoyO-ethylJ-benzenesulfonylJ-aminoJ-propionic acid;
{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonylaminoj-acetic acid;
4-{4-[2-Cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonylaminoj-butyric acid;
(Carboxymethyl-{4-[2-cyclopentyl-1-(5-methoxy-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)- ethyl]-benzenesulfonyl}-amino)-acetic acid;
3-[{4-[2-Cyclopentyl-1-(5-trifluoromethyl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{2-Chloro-4-[1-(5-chloro-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-2-cyclopentyl-ethyl]- benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid;
3-[{4-[2-Cyclopentyl-1-(5-ethyl-thiazolo[5,4-b]pyridin-2-ylcarbamoyl)-ethyl]-2- trifluoromethyl-benzenesulfonyl}-(2-methoxy-ethyl)-amino]-propionic acid.
PCT/US2006/038201 2005-09-30 2006-09-28 Sulfonamide derivatives as glycokinase activators useful in the treatment of type 2 diabetes WO2007041366A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
CA002623045A CA2623045A1 (en) 2005-09-30 2006-09-28 Sulfonamide derivatives as glycokinase activators useful in the treatment of type 2 diabetes
US12/088,594 US8252931B2 (en) 2005-09-30 2006-09-28 Thiazolo[5,4-B]pyridine glucokinase activators
EP06815877A EP1934233B1 (en) 2005-09-30 2006-09-28 Sulfonamide derivatives as glycokinase activators useful in the treatment of type 2 diabetes
BRPI0616710-1A BRPI0616710A2 (en) 2005-09-30 2006-09-28 sulfonamide derivatives as glycokinase activators usable in the treatment of type 2 diabetes
JP2008533703A JP2009510111A (en) 2005-09-30 2006-09-28 Sulfonamide derivatives as glucokinase activators useful in the treatment of type 2 diabetes
AU2006297128A AU2006297128B2 (en) 2005-09-30 2006-09-28 Sulfonamide derivatives as glycokinase activators useful in the treatment of type 2 diabetes
ES06815877T ES2385052T3 (en) 2005-09-30 2006-09-28 Sulfonamide derivatives as glucokinase activators useful in the treatment of type 2 diabetes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US72263005P 2005-09-30 2005-09-30
US60/722,630 2005-09-30

Publications (1)

Publication Number Publication Date
WO2007041366A1 true WO2007041366A1 (en) 2007-04-12

Family

ID=37606931

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/038201 WO2007041366A1 (en) 2005-09-30 2006-09-28 Sulfonamide derivatives as glycokinase activators useful in the treatment of type 2 diabetes

Country Status (15)

Country Link
US (1) US8252931B2 (en)
EP (2) EP2298779A1 (en)
JP (1) JP2009510111A (en)
KR (1) KR20080056175A (en)
CN (1) CN101273048A (en)
AR (1) AR056109A1 (en)
AU (1) AU2006297128B2 (en)
BR (1) BRPI0616710A2 (en)
CA (1) CA2623045A1 (en)
ES (1) ES2385052T3 (en)
GT (1) GT200600428A (en)
PE (1) PE20070489A1 (en)
RU (1) RU2419624C2 (en)
TW (1) TW200800896A (en)
WO (1) WO2007041366A1 (en)

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008017381A1 (en) 2006-08-08 2008-02-14 Sanofi-Aventis Arylaminoaryl-alkyl-substituted imidazolidine-2,4-diones, processes for preparing them, medicaments comprising these compounds, and their use
WO2008119734A2 (en) * 2007-03-29 2008-10-09 Novartis Ag Process for the manufacture of organic compounds
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
WO2009091014A1 (en) 2008-01-18 2009-07-23 Astellas Pharma Inc. Phenyl acetamide derivative
WO2010003624A2 (en) 2008-07-09 2010-01-14 Sanofi-Aventis Heterocyclic compounds, processes for their preparation, medicaments comprising these compounds, and the use thereof
WO2010068601A1 (en) 2008-12-08 2010-06-17 Sanofi-Aventis A crystalline heteroaromatic fluoroglycoside hydrate, processes for making, methods of use and pharmaceutical compositions thereof
US7741327B2 (en) 2008-04-16 2010-06-22 Hoffmann-La Roche Inc. Pyrrolidinone glucokinase activators
US7750020B2 (en) 2004-04-02 2010-07-06 Novartis Ag Sulfonamide-thiazolpyridine derivatives as glucokinase activators useful the treatment of Type 2 diabetes
US7781451B2 (en) 2004-04-02 2010-08-24 Novartis Ag Thiazolopyridine derivatives, pharmaceut ical conditions containing them and methods of treating glucokinase mediated conditions
US7795257B2 (en) 2005-09-30 2010-09-14 Novartis Ag Organic compounds
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
EP2280001A1 (en) * 2008-04-24 2011-02-02 Banyu Pharmaceutical Co., Ltd. Long-chain fatty acid elongation enzyme inhibitor comprising arylsulfonyl derivative as active ingredient
WO2011023754A1 (en) 2009-08-26 2011-03-03 Sanofi-Aventis Novel crystalline heteroaromatic fluoroglycoside hydrates, pharmaceuticals comprising these compounds and their use
US7935699B2 (en) 2006-07-24 2011-05-03 Hoffmann-La Roche Inc. Pyrazole glucokinase activators
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
US8222416B2 (en) 2009-12-14 2012-07-17 Hoffmann-La Roche Inc. Azaindole glucokinase activators
US8252931B2 (en) 2005-09-30 2012-08-28 Novartis Ag Thiazolo[5,4-B]pyridine glucokinase activators
US8258134B2 (en) 2008-04-16 2012-09-04 Hoffmann-La Roche Inc. Pyridazinone glucokinase activators
WO2012120052A1 (en) 2011-03-08 2012-09-13 Sanofi Oxathiazine derivatives substituted with carbocycles or heterocycles, method for producing same, drugs containing said compounds, and use thereof
WO2012120054A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120053A1 (en) 2011-03-08 2012-09-13 Sanofi Branched oxathiazine derivatives, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120055A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120056A1 (en) 2011-03-08 2012-09-13 Sanofi Tetrasubstituted oxathiazine derivatives, method for producing them, their use as medicine and drug containing said derivatives and the use thereof
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013057944A1 (en) 2011-10-19 2013-04-25 興和株式会社 Novel spiroindoline compound, and medicinal agent comprising same
US8946440B2 (en) 2008-04-28 2015-02-03 Kyorin Pharmaceutical Co., Ltd. Cyclopentylacrylamide derivative

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW200831081A (en) 2006-12-25 2008-08-01 Kyorin Seiyaku Kk Glucokinase activator
KR20090121376A (en) * 2007-03-07 2009-11-25 교린 세이야꾸 가부시키 가이샤 Glucokinase activator
EP2283349A1 (en) * 2008-04-29 2011-02-16 Becton, Dickinson & Company Method and system for measuring a sample to determine the presence of and optionally treat a pathologic condition
CN104592073B (en) * 2015-02-12 2016-03-30 佛山市赛维斯医药科技有限公司 One class is containing N-diamantane amides, the purposes of alcoxyl benzene
CN104610112B (en) * 2015-02-12 2016-06-01 佛山市赛维斯医药科技有限公司 N-phenyl diamantane amides glucokinase activators, preparation method and its usage
CN104610113B (en) * 2015-02-12 2016-02-17 佛山市赛维斯医药科技有限公司 A kind of N-phenyl diamantane amides of nitrile group-containing benzene and purposes
CN104610115B (en) * 2015-02-12 2016-03-16 佛山市赛维斯医药科技有限公司 A kind of N-phenyl diamantane amides containing p-nitrophenyl and purposes
CN104693085B (en) * 2015-02-12 2016-06-01 佛山市赛维斯医药科技有限公司 Containing N-phenyl diamantane amides, the Its Preparation Method And Use of halogeno-benzene
CN104610114B (en) * 2015-02-12 2016-06-01 佛山市赛维斯医药科技有限公司 The N-diamantane amides that one class alkoxyphenyl radical replaces and purposes thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
WO2004072031A2 (en) 2003-02-11 2004-08-26 Prosidion Limited Phenylacetamides and their use as glucokinase modulators
WO2005095418A1 (en) 2004-04-02 2005-10-13 Novartis Ag Sulfonamide-thiazolpyridine derivatives as glucokinase activators useful the treatment of type 2 diabetes

Family Cites Families (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6706406B1 (en) * 1996-11-13 2004-03-16 Fern Investments Limited Composite steel structural plastic sandwich plate systems
RU2242469C2 (en) * 1999-03-29 2004-12-20 Ф.Хоффманн-Ля Рош Аг Glucokinase activating agents
MXPA01009814A (en) * 1999-03-29 2002-04-24 Hoffmann La Roche Glucokinase activators.
US6320050B1 (en) * 1999-03-29 2001-11-20 Hoffmann-La Roche Inc. Heteroaromatic glucokinase activators
US6610846B1 (en) * 1999-03-29 2003-08-26 Hoffman-La Roche Inc. Heteroaromatic glucokinase activators
US6353111B1 (en) * 1999-12-15 2002-03-05 Hoffmann-La Roche Inc. Trans olefinic glucokinase activators
CA2407416C (en) 2000-05-03 2006-07-18 Paige Erin Mahaney Alkynyl phenyl heteroaromatic glucokinase activators
ES2248309T3 (en) * 2000-05-03 2006-03-16 F. Hoffmann-La Roche Ag GLUCOQUINASE ACTIVATORS CONTAINING HYDANTOIN.
ES2230309T3 (en) 2000-05-08 2005-05-01 F. Hoffmann-La Roche Ag PHENYLACETAMIDES REPLACED AND ITS EMPLOYMENT WITH QUCOKINASA ACTIVITIES.
AU6591401A (en) 2000-05-08 2001-11-20 Hoffmann La Roche Para-amine substituted phenylamide glucokinase activators
US6489485B2 (en) * 2000-05-08 2002-12-03 Hoffmann-La Roche Inc. Para-amine substituted phenylamide glucokinase activators
ES2243547T3 (en) * 2000-07-20 2005-12-01 F. Hoffmann-La Roche Ag BENCENACETAMIDE REPLACED FOR ALFA-ACILO AND ALFA HETEROATOMOS AS A GLUCOKINASE ACTIVATOR.
US6369232B1 (en) 2000-08-15 2002-04-09 Hoffmann-La Roche Inc. Tetrazolyl-phenyl acetamide glucokinase activators
FR2816509B1 (en) * 2000-11-15 2004-02-06 Sod Conseils Rech Applic ASSOCIATION OF CALPAIN INHIBITORS AND OXYGEN REACTIVE TRAPS
KR100545431B1 (en) * 2000-12-06 2006-01-24 에프. 호프만-라 로슈 아게 Fused heteroaromatic glucokinase activators
US6433188B1 (en) * 2000-12-06 2002-08-13 Wendy Lea Corbett Fused heteroaromatic glucokinase activators
US6482951B2 (en) 2000-12-13 2002-11-19 Hoffmann-La Roche Inc. Isoindolin-1-one glucokinase activators
SE0102764D0 (en) 2001-08-17 2001-08-17 Astrazeneca Ab Compounds
US6911545B2 (en) 2001-12-19 2005-06-28 Hoffman-La Roche Inc. Crystals of glucokinase and methods of growing them
CZ2004747A3 (en) 2001-12-21 2004-11-10 Novo Nordisk A/S Amide derivatives functioning as GK activators
EP1496052B1 (en) 2002-03-26 2009-08-05 Banyu Pharmaceutical Co., Ltd. Novel aminobenzamide derivative
CA2482346C (en) 2002-04-26 2010-03-09 F. Hoffmann-La Roche Ag Substituted phenylacetamides and their use as glucokinase activators
AU2003235307A1 (en) 2002-05-16 2003-12-02 Banyu Pharmaceutical Co., Ltd. Crystal of glucokinase protein, and method for drug design using the crystal
JP4881559B2 (en) 2002-06-27 2012-02-22 ノボ・ノルデイスク・エー/エス Arylcarbonyl derivatives as therapeutic agents
AU2003270285B2 (en) * 2002-10-03 2007-04-26 F. Hoffmann-La Roche Ag Indole-3-carboxamides as glucokinase (GK) activators
MY141521A (en) 2002-12-12 2010-05-14 Hoffmann La Roche 5-substituted-six-membered heteroaromatic glucokinase activators
WO2004063194A1 (en) 2003-01-06 2004-07-29 Eli Lilly And Company Heteroaryl compounds
DK1585739T3 (en) 2003-01-06 2011-06-20 Lilly Co Eli Substituted arylcyclopropylacetamides as glucokinase activators
JP4621198B2 (en) * 2003-02-11 2011-01-26 プロシディオン・リミテッド Tri (cyclo) substituted amide glucokinase activating compound
AU2004220234C1 (en) 2003-02-13 2013-01-17 Msd K.K. Novel 2-pyridinecarboxamide derivatives
EP1600442B1 (en) 2003-02-26 2018-01-17 Msd K.K. Heteroarylcarbamoylbenzene derivative
CA2561157A1 (en) * 2004-04-02 2005-10-13 Novartis Ag Thiazolopyridine derivatives, pharmaceutical conditions containing them and methods of treating glucokinase mediated conditions
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
JP2008521864A (en) 2004-12-03 2008-06-26 トランステック・ファーマ、インコーポレイテッド Heteroaromatic glucokinase activator
GT200600428A (en) 2005-09-30 2007-05-21 ORGANIC COMPOUNDS
GT200600429A (en) * 2005-09-30 2007-04-30 ORGANIC COMPOUNDS

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
WO2004072031A2 (en) 2003-02-11 2004-08-26 Prosidion Limited Phenylacetamides and their use as glucokinase modulators
WO2005095418A1 (en) 2004-04-02 2005-10-13 Novartis Ag Sulfonamide-thiazolpyridine derivatives as glucokinase activators useful the treatment of type 2 diabetes

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BERRY ET AL., J. CELL BIOL., vol. 43, 1969, pages 506 - 520
GREENE; WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY AND SONS, INC.
MCOMIE: "Protective Groups in Organic Chemistry", 1973, PLENUM PRESS
PATEL MONA, EXPERT OPIN INVESTIG DRUGS, vol. 12, no. 4, 2003, pages 623 - 633

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US7750020B2 (en) 2004-04-02 2010-07-06 Novartis Ag Sulfonamide-thiazolpyridine derivatives as glucokinase activators useful the treatment of Type 2 diabetes
US7781451B2 (en) 2004-04-02 2010-08-24 Novartis Ag Thiazolopyridine derivatives, pharmaceut ical conditions containing them and methods of treating glucokinase mediated conditions
US8252931B2 (en) 2005-09-30 2012-08-28 Novartis Ag Thiazolo[5,4-B]pyridine glucokinase activators
US7795257B2 (en) 2005-09-30 2010-09-14 Novartis Ag Organic compounds
US7935699B2 (en) 2006-07-24 2011-05-03 Hoffmann-La Roche Inc. Pyrazole glucokinase activators
WO2008017381A1 (en) 2006-08-08 2008-02-14 Sanofi-Aventis Arylaminoaryl-alkyl-substituted imidazolidine-2,4-diones, processes for preparing them, medicaments comprising these compounds, and their use
WO2008119734A3 (en) * 2007-03-29 2009-01-15 Novartis Ag Process for the manufacture of organic compounds
WO2008119734A2 (en) * 2007-03-29 2008-10-09 Novartis Ag Process for the manufacture of organic compounds
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
WO2009091014A1 (en) 2008-01-18 2009-07-23 Astellas Pharma Inc. Phenyl acetamide derivative
US8329707B2 (en) 2008-01-18 2012-12-11 Astellas Pharma Inc. Substituted pyrazine compounds
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
EP2280001A1 (en) * 2008-04-24 2011-02-02 Banyu Pharmaceutical Co., Ltd. Long-chain fatty acid elongation enzyme inhibitor comprising arylsulfonyl derivative as active ingredient
EP2280001A4 (en) * 2008-04-24 2012-01-18 Msd Kk Long-chain fatty acid elongation enzyme inhibitor comprising arylsulfonyl derivative as active ingredient
US8420823B2 (en) 2008-04-24 2013-04-16 Msd K.K. Long-chain fatty acyl elongase inhibitor comprising arylsulfonyl derivative as active ingredient
US9452977B2 (en) 2008-04-28 2016-09-27 Kyorin Pharmaceutical Co., Ltd. Cyclopentylacrylamide derivative
US8946440B2 (en) 2008-04-28 2015-02-03 Kyorin Pharmaceutical Co., Ltd. Cyclopentylacrylamide derivative
WO2010003624A2 (en) 2008-07-09 2010-01-14 Sanofi-Aventis Heterocyclic compounds, processes for their preparation, medicaments comprising these compounds, and the use thereof
WO2010068601A1 (en) 2008-12-08 2010-06-17 Sanofi-Aventis A crystalline heteroaromatic fluoroglycoside hydrate, processes for making, methods of use and pharmaceutical compositions thereof
WO2011023754A1 (en) 2009-08-26 2011-03-03 Sanofi-Aventis Novel crystalline heteroaromatic fluoroglycoside hydrates, pharmaceuticals comprising these compounds and their use
US8222416B2 (en) 2009-12-14 2012-07-17 Hoffmann-La Roche Inc. Azaindole glucokinase activators
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012120053A1 (en) 2011-03-08 2012-09-13 Sanofi Branched oxathiazine derivatives, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120055A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120056A1 (en) 2011-03-08 2012-09-13 Sanofi Tetrasubstituted oxathiazine derivatives, method for producing them, their use as medicine and drug containing said derivatives and the use thereof
WO2012120054A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120052A1 (en) 2011-03-08 2012-09-13 Sanofi Oxathiazine derivatives substituted with carbocycles or heterocycles, method for producing same, drugs containing said compounds, and use thereof
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013057944A1 (en) 2011-10-19 2013-04-25 興和株式会社 Novel spiroindoline compound, and medicinal agent comprising same

Also Published As

Publication number Publication date
AU2006297128B2 (en) 2010-12-23
EP1934233B1 (en) 2012-05-16
CN101273048A (en) 2008-09-24
PE20070489A1 (en) 2007-06-13
ES2385052T3 (en) 2012-07-17
CA2623045A1 (en) 2007-04-12
US8252931B2 (en) 2012-08-28
RU2419624C2 (en) 2011-05-27
KR20080056175A (en) 2008-06-20
BRPI0616710A2 (en) 2011-06-28
EP1934233A1 (en) 2008-06-25
RU2008116581A (en) 2009-11-10
JP2009510111A (en) 2009-03-12
AR056109A1 (en) 2007-09-19
AU2006297128A1 (en) 2007-04-12
GT200600428A (en) 2007-05-21
EP2298779A1 (en) 2011-03-23
TW200800896A (en) 2008-01-01
US20080312256A1 (en) 2008-12-18

Similar Documents

Publication Publication Date Title
US8252931B2 (en) Thiazolo[5,4-B]pyridine glucokinase activators
AU2006297127B2 (en) 3-cyclyl-2- (4-sulfamo yl-phenyl) -n-cyclyl-propionamide derivatives useful in the treatment of impaired glucose tolerance and diabetes
AU2005229415B2 (en) Thiazolopyridine derivatives, pharmaceutical conditions containing them and methods of treating glucokinase mediated conditions
JP4700684B2 (en) Sulfonamide-thiazolopyridine derivatives as glucokinase activators useful in the treatment of type 2 diabetes
US7812167B2 (en) Substituted (thiazol-2-yl)-amides or sulfonamides as glucokinase activators useful in the treatment of type 2 diabetes

Legal Events

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

Ref document number: 200680035484.7

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2006815877

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1208/DELNP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2006297128

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2623045

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/004179

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2008533703

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12088594

Country of ref document: US

Ref document number: 1020087007627

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2006297128

Country of ref document: AU

Date of ref document: 20060928

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2008116581

Country of ref document: RU

ENP Entry into the national phase

Ref document number: PI0616710

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080328