WO2004046117A1 - Pyridazinone derivatives as gsk-3beta inhibitors - Google Patents

Pyridazinone derivatives as gsk-3beta inhibitors Download PDF

Info

Publication number
WO2004046117A1
WO2004046117A1 PCT/EP2003/012950 EP0312950W WO2004046117A1 WO 2004046117 A1 WO2004046117 A1 WO 2004046117A1 EP 0312950 W EP0312950 W EP 0312950W WO 2004046117 A1 WO2004046117 A1 WO 2004046117A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
aryl
oxo
heteroaryl
heterocyclyl
Prior art date
Application number
PCT/EP2003/012950
Other languages
French (fr)
Inventor
Swen Hoelder
Thorsten Naumann
Karl Schoenafinger
David William Will
Hans Matter
Guenter Mueller
Dominique Le Suisse
Bernard Baudoin
Thomas Rooney
Franck Halley
Gilles Tiraboschi
Original Assignee
Aventis Pharma Deutschland 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 Aventis Pharma Deutschland Gmbh filed Critical Aventis Pharma Deutschland Gmbh
Priority to JP2004552660A priority Critical patent/JP2006509748A/en
Priority to CA002506022A priority patent/CA2506022A1/en
Priority to DE60315354T priority patent/DE60315354T2/en
Priority to AU2003283414A priority patent/AU2003283414A1/en
Priority to MXPA05005270A priority patent/MXPA05005270A/en
Priority to BR0316720-8A priority patent/BR0316720A/en
Priority to EP03775372A priority patent/EP1581505B1/en
Publication of WO2004046117A1 publication Critical patent/WO2004046117A1/en
Priority to NO20052887A priority patent/NO20052887L/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/26Psychostimulants, e.g. nicotine, cocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • 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
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/02Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings
    • C07D237/06Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D237/10Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-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
    • C07D237/14Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/02Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings
    • C07D237/06Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D237/10Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-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
    • C07D237/24Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D417/14Heterocyclic 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 three or more hetero rings
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/582Recycling of unreacted starting or intermediate materials

Definitions

  • the present invention relates to compounds according to the general formula (I), with the definitions of the substituents A and Ar given below in the text, as well as their physiologically acceptable salts, methods for producing these compounds and their use as pharmaceuticals.
  • These compounds are kinase inhibitors, in particular inhibitors of the kinase GSK- 3 ⁇ (glycogen synthase kinase-3 ⁇ ).
  • Pyridazinone derivatives are well known pharmaceuticals, but it has not been reported so far that pyridazinone derivatives can be employed for the inhibition of GSK-3 ⁇ or tau-phosporylation, respectively. Pyridazinone derivatives described in literature differ from those of the present invention due to a different substitution pattern and (partially) different indications.
  • WO 03/059891 discloses pyridazinone derivatives that are useful for treating diseases and conditions caused or exacerbated by unregulated p38 MAP Kinase activity and/or TNF activity.
  • the compounds described therein can be used, for example, for the treatment of inflammatory conditions, diabetes, Alzheimer's disease or cancer. They differ from the compounds of the present invention in the substitution of the pyridazinone cycle, since the nitrogen at position 2 of the cycle is mostly substituted with alky, aryl or heteroaryl and at position 4 of the cycle there is no amido group defined as substituent (equals substituent A of the compounds of the present invention).
  • the object of the present invention is to provide compounds showing this ability.
  • A represents A1 or A2
  • R is unsubstituted or at least monosubstituted Ci-Cio-alkyl, aryl, aryl-(C ⁇ -C ⁇ 0 - alkyl)-, heteroaryl, heteroaryl-(C ⁇ -C ⁇ o-alkyl)-, heterocyclyl, heterocyclyl-(Cr C 10 -alkyl)-, C 3 -C ⁇ 0 -cycloalkyl, polycycloalkyl, C 2 -C ⁇ 0 -alkenyl or C 2 -C ⁇ 0 - alkinyl,
  • substituents are selected from halogen, -CN, C C ⁇ 0 -alkyl, -N0 2 , - OR1 , -C(0)OR1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -SR1 , - S(0)R1 , -S0 2 R1 , -NHS0 2 R1 , -S0 2 NR1 R2, -C(S)NR1 R2, -NHC(S)R1 ,
  • aryl, heterocyclyl and heteroaryl may in turn be at least monosubstituted with Ci-C ⁇ -alkyl, Ci-C ⁇ -alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
  • Ar is unsubstituted or at least monosubstituted aryl or heteroaryl
  • substituents are selected from halogen, -CN, NO 2 , CrC ⁇ 0 -alkyl, -OR1 , -C(0)OR1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -NHC(S)R1 , -C(S)NR1 R2, -SR1 , -S(0)R1 , -S0 2 R1 , -NHS0 2 R1 , -S0 2 NR1 R2, -0-S0 2 R1 , -SO 2 -O-RI , aryl, heteroaryl, ary CrC-e-alkyl)-, formyl, trifluoromethyl and trifluoromethoxy,
  • aryl and heteroaryl may in turn be at least monosubstituted with C ⁇ -C 6 - alkyl, CrC ⁇ -alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
  • R1 and R2 are independently from each other
  • heteroaryl is a 5 to 10-membered, aromatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O und S;
  • aryl is phenyl, indanyl, indenyl or naphthyl
  • heterocyclyl is a 5 to 10-membered, aliphatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O and S;
  • A is not -C(0)NH(C ⁇ -C ⁇ -alkyl)
  • Ar is phenyl which is at least monosubstituted with heterocyclyl or heteroaryl containing nitrogen.
  • groups, fragments, residues or substituents such as, for example, aryl, heteroaryl, alkyl etc., may be present several times, they all independently from each other have the meanings indicated and may hence, in each individual case, be identical with or different from each other.
  • aryl as well as to any other residue independently from its classification as aryl group, -substituent, -fragment or - residue.
  • di(CrCi 0 -alkyl)amino group in which the alkyl substitutents may be identical or different (for instance 2 x ethyl or 1 x propyl and 1 x heptyl).
  • a substituent for example aryl
  • a substituent may be unsubstituted or at least mono-substituted with a group of further substituents, for example, C ⁇ -C 6 -alkyl, C ⁇ -C 6 -alkoxy, halogen etc.
  • a group of further substituents for example, C ⁇ -C 6 -alkyl, C ⁇ -C 6 -alkoxy, halogen etc.
  • aryl may be substituted twice with ethyl, aryl may be mono-substituted with methyl or ethoxy, respectively, aryl may be mono-substituted with ethyl or fluoro, respectively, aryl may be substituted twice with methoxy, etc.
  • Alkyl, alkenyl and alkynyl residues may be linear or branched. This also applies when they are part of other groups, for example in alkoxy groups (C C ⁇ 0 -alkyl-O-), alkoxycarbonyl groups or amino groups, or when they are substituted.
  • alkyl groups are: methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl. This comprises both the n-isomers of these residues and isopropyl, isobutyl, isopentyl, sec-butyl, tert-butyl, neopentyl, 3,3-dimethylbutyl etc.
  • alkyl here also includes unsubstituted alkyl residues as well as alkyl residues which are substituted by one or more, for example one, two, three or four, identical or different residues, for example aryl, heteroaryl, alkoxy or halogen.
  • the substituents may be present in any desired position of the alkyl group.
  • cycloalkyl residues are: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. All cycloalkyl groups may be unsubstituted or optionally substituted by one or more further residues, as exemplified above in the case of the alkyl groups.
  • alkenyl and alkynyl groups are the vinyl residue, the 1-propenyl residue, the 2-propenyl residue (allyl residue), the 2-butenyl residue, the 2-methyl- 2-propenyl residue, the 3-methyl-2-butenyl residue, the ethynyl residue, the 2- propynyl residue (propargyl residue), the 2-butynyl residue or the 3-butynyl residue.
  • alkenyl here also expressly includes cycloalkenyl residues and cycloalkenyl-alkyl-residues (alkyl substituted by cycloalkenyl) containing at least three carbon atoms.
  • Examples for cycloalkenyl residues are cyclopentenyl, cyclohexenyl, cycloheptenyl and cyclooctenyl.
  • the alkenyl residues may have 1 to 3 conjugated or unconjugated double bonds in a straight or branched chain; the same applies to alkynyl residues in respect of triple bonds.
  • the alkenyl and alkinyl residues may be unsubstituted or optionally substituted by one or more further residues, as exemplified above in the case of the alkyl groups.
  • polycycloalkyl residues examples are: adamantyl, quinuclidinyl, bornanyl, norbornanyl, bornenyl and norbornenyl. If not stated otherwise, the above-mentioned aryl, heteroaryl and heterocyclic residues may be unsubstituted or may carry one or more, for example one, two, three or four of the substituents indicated in the above definition, which substituents may be in any desired position.
  • the substituent may be in the 2-position, the 3-position or the 4- position
  • the substituents in disubstituted phenyl residues the substituents may be in 2,3-position, 2,4-position, 2,5-position, 2,6-position, 3,4-position or 3,5-position.
  • the substituents In trisubstituted phenyl residues the substituents may be in 2,3,4-position, 2,3,5-position, 2,3,6- position, 2,4,5-position, 2,4,6-position or 3,4,5-position.
  • the substituents In fourfold substituted phenyl residues, the substituents may be in the 2,3,4,5-position, the 2,3,4,6- position, or the 2, 3,5,6-position.
  • divalent residues may be attached to the adjacent groups by any ring carbon atom.
  • a phenylene residue these may be in 1 , 2-position (ortho-phenylene), 1 , 3-position (meta-phenylene) or 1 ,4-position (para-phenylene).
  • 5-membered ring aromatics containing one heteroatom such as, for example, thiophene or furan
  • the two free bonds may be in 2,3-position, 2,4-position, 2,5-position or 3,4-position.
  • a divalent residue derived from pyridine may be a 2,3-, 2,4-, 2,5-, 2,6-, 3,4- or 3,5-pyridinediyl residue.
  • the present invention includes all positional isomers, i. e., in the case of a 2,3-pyridinediyl residue, for example, it includes the compound in which the one adjacent group is present in the 2-position and the other adjacent group is present in the 3-position as well as the compound in which the one adjacent group is present in the 3-position and the other adjacent group is present in the 2-position.
  • heteroaryl residues, heteroarylene residues, heterocyclyl residues, heterocyclylen residues and rings which are formed by two groups bonded to a nitrogen are preferably derived from completely saturated, partially saturated or completely unsaturated heterocycles (i.e. heterocycloalkanes, heterocycloalkenes, heteroaromatics), which contain one, two, three or four heteroatoms, which may be identical or different; more preferably they are derived from heterocycles which contain one, two, or three, in particular one or two, heteroatoms, which may be identical or different.
  • the heterocycles may be monocyclic or polycyclic, for example monocyclic, bicyclic or tricyclic.
  • the rings Preferably they are monocyclic or bicyclic.
  • the rings preferably are 5- membered rings, 6-membered rings or 7-membered rings.
  • aryl and the term “heteroaryl” as used herein comprise bicyclic residues in which both cycles are aromatic as well as bicyclic residues in which only one cycle is aromatic.
  • Suitable aliphatic heterocycles include, for example, the saturated heterocycles pyrrolidine, piperidine, piperazine, imidazolidine, pyrazolidine, isothiazolidine, thiazolidine, isoxazolidine, oxazolidine, tetrahydrofuran, dioxolane, 2-oxo-azepane, morpholine and thiomorpholine as well as the partially unsaturated heterocycles isochromamyl, chromamyl, 1 ,2,3,4-tetrahydroisochinolyl and 1 ,2,3,4- tetrahydrochinolyl.
  • the degree of saturation of heterocyclic groups is indicated in their individual definitions.
  • Substituents which may be derived from these heterocycles may be attached via any suitable carbon atom.
  • Residues derived from nitrogen heterocycles may carry a hydrogen atom or a substituent on a ring nitrogen atom, and examples include pyrrole, imidazole, pyrrolidine, morpholine, piperazine residues, etc.
  • Those nitrogen heterocyclic residues may also be attached via a ring nitrogen atom, in particular if the respective heterocyclic residue is bonded to a carbon atom.
  • Suitable nitrogen heterocycles may also be present as N-oxides or as quarternary salts containing a counterion which is derived from a physiologically acceptable acid.
  • Pyridyl residues for example, may be present as pyridine-N-oxides.
  • Arylalkyl means an alkyl residue, which in turn is substituted by an aryl residue.
  • Heteroarylalkyl means an alkyl residue, which in turn is substituted by a heteroaryl residue.
  • Heterocyclylalkyl means an alkyl residue, which in turn is substituted by a heterocyclyl residue.
  • alkyl, heteroaryl, heterocyclyl and aryl it is referred to the above-mentioned definitions.
  • Halogen is fluorine, chlorine, bromine or iod, preferably fluorine, chlorine or bromine, most preferably fluorine or chlorine.
  • the present invention includes all stereoisomeric forms of the compounds of the formula (I). Centers of asymmetry that are present in the compounds of formula (I) all independently of one another have S configuration or R configuration.
  • the invention includes all possible enantiomers and diastereomers and mixtures of two or more stereoisomers, for example mixtures of enantiomers and/or diastereomers, in all ratios.
  • compounds according to the present invention which may exist as enantiomers may be present in enantiomerically pure form, both as levorotatory and as dextrorotatory antipodes, in the form of racemates and in the form of mixtures of the two enantiomers in all ratios.
  • the invention includes both the cis form and the trans form as well as mixtures of these forms in all ratios. All these forms are an object of the present invention.
  • the preparation of individual stereoisomers may be carried out, if desired, by separation of a mixture by customary methods, for example by chromatography or crystallization, by the use of stereochemically uniform starting materials for the synthesis or by stereoselective synthesis.
  • a derivatization may be carried out before a separation of stereoisomers.
  • the separation of a mixture of stereoisomers may be carried out at the stage of the compounds of the formula (I) or at the stage of an intermediate during the synthesis.
  • the present invention also includes all tautomeric forms of the compounds of formula (I), in particular keto-enol tautomehsm, i.e. the respective compounds may be present either in their keto form or in their enol form or in mixtures thereof in all ratios.
  • the invention also comprises their corresponding physiologically or toxicologically acceptable salts.
  • Physiologically acceptable salts are particularly suitable for medical applications, due to their greater solubility in water compared with the starting or base compounds. Said salts must have a physiologically acceptable anion or cation.
  • Suitable physiologically acceptable acid addition salts of the compounds of the invention are salts of inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, metaphosphoric acid, nitric acid, sulfonic acid and sulfuric acid and also of organic acids such as, for example, acetic acid, theophyllinacetic acid, methylene-bis-b-oxynaphthoic acid, benzenesulfonic acid, benzoic acid, citric acid, ethanesulfonic acid, salicylic acid, fumaric acid, gluconic acid, glycolic acid, isethionic acid, lactic acid, lactobionic acid, maleic acid, malic acid, methane- sulfonic acid, succinic acid, p-toluenesulfonic acid, tartaric acid and trifluoroacetic acid.
  • Suitable pharmaceutically acceptable basic salts are ammonium salts, alkali metal salts (such as sodium salts and potassium salts
  • Salts having a pharmaceutically unacceptable anion are likewise included within the scope of the present invention as useful intermediates for preparing or purifying pharmaceutically acceptable salts and/or for use in nontherapeutic applications, for example in-vitro applications.
  • the invention also includes, in addition to the salt forms mentioned, inner salts or betaines (zwitterions).
  • the respective salts according to the formula (I) may be obtained by customary methods which are known to the person skilled in the art like, for example by contacting these with an organic or inorganic acid or base in a solvent or dispersant, or by anion exchange or cation exchange with other salts.
  • the present invention furthermore includes all solvates of compounds of the formula (I), for example hydrates or adducts with alcohols, active metabolites of the compounds of the formula (I), and also derivatives, which contain physiologically tolerable and cleavable groups, for example esters or amides.
  • physiologically functional derivative used herein relates to any physiologically acceptable derivative of an inventive compound of the formula I, for example an ester which on administration to a mammal, for example humans, is capable of forming (directly or indirectly) a compound of the formula I or an active metabolite thereof.
  • the physiologically functional derivatives also include prodrugs of the compounds of the invention.
  • prodrugs may be metabolized in vivo to a compound of the invention.
  • These prodrugs may or may not be active themselves and are also object of the present invention.
  • the compounds of the invention may also be present in various polymorphous forms, for example as amorphous and crystalline polymorphous forms. All polymorphous forms of the compounds of the invention are included within the scope of the invention and are another aspect of the invention.
  • A is A1 ;
  • R is unsubstituted or at least monosubstituted CrCio-alkyl, aryl, aryl-(C ⁇ -C ⁇ o- alkyl)-, heteroaryl, heteroaryl-(Ci-Cio-alkyl)-, heterocyclyl, heterocyclyl- (C-i- Cio-alkyl)-, C 3 -C ⁇ o-cycloalkyl, polycycloalkyl, C 2 -C ⁇ o-alkenyl or C- 2 -C 10 - alkinyl,
  • substituents are selected from halogen, -CN, d-Cio-alkyl, -NO 2 , -OR1 , -C(0)OR1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -SR1 , -S(0)R1 , -SO2RI , -NHSO2RI . -S0 2 NR1 R2, -C(S)NR1 R2, -NHC(S)R1 ,
  • heterocyclyl and heteroaryl may in turn be at least monosubstituted with C-i-C ⁇ -alkyl, CrC 6 -alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
  • heteroaryl is a 5 to 10-membered, aromatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O and S;
  • aryl is phenyl, indanyl, indenyl or naphthyl
  • heterocyclyl is a 5 to 10-membered, aliphatic, mono- Oder bicyclic heterocycle containing one or more heteroatoms selected from N, O and S;
  • R is unsubstituted or at least monosubstituted Ci-Cio-alkyl, aryl, aryl-(C ⁇ -C ⁇ o- alkyl)-, heterocyclyl, heterocyclyl-(CrCi 0 -alkyl)-, C 3 -C ⁇ . 0 -cycloalkyl, heteroaryl or heteroaryl-(CrC ⁇ o-alkyl)-,
  • substituents are selected from halogen, -CN, C ⁇ -C ⁇ 0 -Alkyl, -NO 2 , -OR1 , -C(0)OR1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -SR1 , -S(0)R1 , -SO 2 RI , -NHSO 2 RI , -S0 2 NR1 R2, -C(S)NR1 R2, -NHC(S)R1 , -O-SO 2 RI , -SO2-O-RI , oxo, -C(0)R1 , -C(NH)NH 2 , heterocyclyl, C 3 -C 10 - cycloalkyl, aryl-(d ⁇ C 6 -alkyl)-, aryl, heteroaryl, trifluoromethyl, trifluoromethylsulfanyl and trifluoromethoxy,
  • heterocyclyl and heteroaryl may in turn be at least monosubstituted with CrC ⁇ -alkyl, d-C- ⁇ -alkoxy, halogen, trifluoromethyl, trifluoroethoxy or OH;
  • R1 and R2 are independently from each other
  • heteroaryl is a 5 to 10-membered, aromatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O and S;
  • aryl is phenyl, indanyl, indenyl or naphthyl
  • heterocyclyl is a 5 to 10-membered, aliphatic, mono- Oder bicyclic heterocycle, containing one or more heteroatoms selected from N, 0 and S;
  • Ar is unsubstituted or at least monosubstituted phenyl, pyridinyl, pyrimidinyl, pyrazolyl, thiophenyl, isoxaloyl, benzo[b]thiophenyl, benzodioxolyl or thiazolo[3,2-b][1 ,2,4]-tiazolyl,
  • substituents are selected from halogen, -CN, N0 2 , CrCio-alkyl, -OR1 , -C(0)0R1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -NHC(S)R1 , -C(S)NR1 R2, -SR1 , -S(0)R1 , -S0 2 R1 , -NHS0 2 R1 , -SO 2 NRI R2, -O-SO 2 RI , -SO 2 -O-RI , aryl, heteroaryl, aryl-(d-C 6 -alkyl)-, formyl, trifluoromethyl and trifluoromethoxy,
  • aryl and heteroaryl may in turn be at least monosubstituted with d-C 6 - alkyl, C ⁇ -C 6 -alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
  • R1 and R2 are independently from each other
  • C Cio-alkyl C 3 -C ⁇ 0 -cycloalkyl, aryl, aryl-(d-C 10 -alkyl)-, C 2 -C ⁇ 0 -alkenyl, C 2 -C ⁇ 0 -alkinyl, heterocyclyl, heterocyclyl-(d-do-alkyl)- or heteroaryl, where the substituents are selected from halogen, C ⁇ -C 6 -alkyl, C C 6 -alkoxy, CN, N0 2 , NH 2 , (C C 6 - alkyl) amino-, di(d-C 6 -alkyl)amino-, OH, COOH, -COO-(d-C 6 -alkyl), -CONH 2 , formyl, trifluoromethyl and trifluoromethoxy;
  • heteroaryl is a 5 to 10-membered aromatic, mono- or bicyclic heterocycle, containing one or more heteroatoms selected from N, O and S;
  • aryl is phenyl, indanyl, indenyl or naphthyl
  • heterocyclyl is a 5 to 10-membered aliphatic, mono- oder bicyclic heterocycle, containing one or more heteroatoms selected from N, O and S;
  • A is A1 ;
  • R is unsubstituted or at least monosubstituted CrCio-alkyl, aryl, aryl-(C ⁇ C ⁇ o- alkyl)-, heterocyclyl, heterocyclyl-(C ⁇ -C ⁇ o-alkyl)-, C 3 -C ⁇ 0 -cycloalkyl, heteroaryl or heteroaryl-(Ci-Cio-alkyl)-,
  • substituents are selected from halogen, CrCio-alkyl, -OR1 , -C(0)OR1 , -NR1 R2, -C(0)NR1 R2, -SR1 , -S0 2 R1 , -S0 2 NR1 R2, oxo, -C(0)R1 , -C(NH)NH 2 , heterocyclyl, C 3 -C 10 -cycloalkyl, aryl-(d-C 6 -alkyl)-, aryl, trifluoromethyl and trifluoromethoxy,
  • heterocyclyl and heteroaryl may in turn be at least monosubtituted with d-C- 6 -alkyl, d-C- ⁇ -alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
  • Ar is unsubstituted or at least monosubstituted phenyl, pyridinyl, pyrimidinyl, pyrazolyl, thiophenyl, isoxazolyl, benzo[b]thiophenyl, benzodioxolyl or thiazolo[3,2-b][1 ,2,4]-tiazolyl,
  • substituents are selected from halogen, Ci-Cio-alkyl, -OR1 , -C(0)OR1 , -NR1 R2, -C(0)NR1 R2, aryl, heteroaryl, aryl-(d-C 6 -alkyl)-, trifluoromethyl and Trifluoromethoxy, and aryl and heteroaryl may in turn be at least monosubsituted with C ⁇ -C 6 - alkyl, d-C 6 -alkoxy, halogen, trifluoromethyl or OH;
  • R1 und R2 are independently from each other
  • d-Cio-alkyl unsubstituted or at least monosubstituted d-Cio-alkyl, C 3 -C ⁇ o ⁇ cycloalkyl, aryl, aryl-(C ⁇ -C ⁇ o-alkyl)-, heterocyclyl, heterocyclyl-(C ⁇ -C ⁇ o-alkyl)- or heteroaryl, where the substituents are selected from halogen, d-C 6 -alkyl, d-Ce-alkoxy, NH 2 , (d-C 6 -alkyl)amino-, di(d-C 6 -alkyl)amino-, OH, trifluoromethyl and trifluoromethoxy;
  • heteroaryl is a 5 to 10-membered, aromatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O und S; heteroaryl is preferably imidazolyl, thiophenyl, furanyl, isoxazolyl, pyridinyl, pyrimidinyl, 1 ,2,3,4- tetrahydrochinolinyl, benzoimidazolyl, indolyl or benzodioxolyl;
  • aryl is phenyl, indanyl, indenyl or naphthyl; aryl is preferably phenyl or naphthyl.
  • heterocyclyl is a 5 to 10-membered, aliphatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O und S; heterocyclyl is preferably 2-oxo-azepanyl, tetrahydrofuranyl, 1 ,3-dioxolanyl, morpholinyl, piperazinyl or piperidinyl;
  • A is A1 ;
  • R is unsubstituted or at least monosubstituted d-Cio-alkyl, aryl, aryl-(C ⁇ -C ⁇ o- alkyl)-, heterocyclyl, heterocyclyl-(d-C 10 -alkyl)-, C 3 -C ⁇ o-cycloalkyl, heteroaryl or heteroaryl-(C ⁇ -C ⁇ o-alkyl)-,
  • substituents are selected from halogen, C Cio-alkyl, -OR1 , -C(0)OR1 , -NR1 R2, -C(0)NR1 R2, -SR1 , -S0 2 R1 , -S0 2 NR1 R2, oxo,
  • -C(0)R1 -C(NH)NH 2 , heterocyclyl, C 3 -C ⁇ 0 -cycloalkyl, aryl-(C C 6 -alkyl)-, aryl, trifluoromethyl and trifluoromethoxy, and aryl, heterocyclyl and heteroaryl may in turn be at least monosubsituted with Ci-Ce-alkyl, C ⁇ -C 6 -alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
  • Ar is unsubstituted or at least monosubstituted phenyl, pyridinyl or pyrimidinyl,
  • substituents are selected from halogen, Ci-Cio-alkyl, -OR1 , -C(0)OR1 , -NR1 R2, -C(0)NR1 R2, aryl, heteroaryl, aryl-(d-C 6 -alkyl)-, trifluoromethyl and trifluoromethoxy,
  • aryl and heteroaryl may in turn be at least monosubsituted with Ci-Ce- alkyl, Ci-C ⁇ -alkoxy, halogen, trifluoromethyl or OH;
  • R1 und R2 are independently from each other
  • Ci-Cio-alkyl unsubstituted or at least monosubstituted Ci-Cio-alkyl, C 3 -do-cycloalkyl, aryl, aryl-(C ⁇ -C ⁇ o-alkyl)-, heterocyclyl, heterocyclyl-(Ci-Cio-alkyl)- or heteroaryl, where the substituents are selected from halogen, Ci-Ce-alkyl, Ci-Ce-alkoxy, NH 2 , (C ⁇ -C 6 -alkyl)amino-, di(d-C 6 -alkyl)amino-, OH, trifluoromethyl and trifluoromethoxy;
  • heteroaryl is a 5 to 10-membered, aromatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O und S; heteroaryl is preferably imidazolyl, thiophenyl, furanyl, isoxazolyl, pyridinyl, pyrimidinyl, 1 ,2,3,4- tetrahydrochinolinyl, benzoimidazolyl, indolyl or benzodioxolyl;
  • aryl is phenyl, indanyl, indenyl or naphthyl; aryl is preferably phenyl or naphthyl.
  • heterocyclyl is a 5 to 10-membered, aliphatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O und S; heterocyclyl is preferably 2-oxo-azepanyl, tetrahydrofuranyl, 1 ,3-dioxolanyl, morpholinyl, piperazinyl or piperidinyl;
  • R is unsubstituted or at least monosubstituted aryl-(C ⁇ -C- 6 -alkyl)- or heteroaryl- (Ci-Ce-alkyl)-,
  • substituents are selected from halogen, C ⁇ -C 6 -alkyl, -OH, -O-aryl, Ci-Ce-alkoxy, -0-(C ⁇ -C 6 -alkylen)-N(C ⁇ -C 6 -alkyl)2, -C(0)OH, -C(0)0-(C ⁇ -C 6 - alkyl), -NH 2 , -N(C ⁇ -C 6 -alkyl) 2 , -NH(C ⁇ -C 6 -alkyl), -NH(d-C 10 -cycloalkyl), -C(0)NH 2 , -C(0)NH-heteroaryl, -C(0)NH-(Ci-C 6 -alkyl), -S0 2 (d-C e -alkyl),
  • aryl, aeterocyclyl and heteroaryl may in turn be at least monosubstituted with d-C 3 -alkyl, C ⁇ -C 3 -alkoxy, fluorine, chlorine, bromine, trifluoromethyl, trifluoromethoxy or OH;
  • heteroaryl is imidazolyl, thiophenyl, furanyl, isoxazolyl, pyridinyl, pyrimidinyl, benzoimidazolyl, indolyl or benzodioxolyl;
  • aryl is phenyl or naphthyl
  • hetrocyclyl is morpholinyl, piperazinyl or piperidinyl;
  • A is A1 ;
  • Ar is unsubstituted or at least monosubstituted phenyl, pyridin-4-yl or pyrimidin-4-yl,
  • substituents are selected from halogen, Ci-Ce-alkyl, -OH, Ci-Ce- alkoxy, -C(0)OH, -C(0)0-(C ⁇ -C 6 -alkyl), -NH 2 , -N(C -C 6 -alkyl) 2 , -NH(d-C 6 - alkyl), -NH(Ci-Cio-cycloalkyl), -NH(heterocyclyl-(d-C 6 -alkyl-)), -NH(aryl-
  • C ⁇ -C 6 -alkyl- (C ⁇ -C 6 -alkyl-)), -C(0)NH 2 , -C(0)NH-(d-C 6 -alkyl), aryl, and heteroaryl, and aryl, heterocyclyl and heteroaryl may in turn be at least monosubstituted C ⁇ -C 3 -Alkyl, d-C 3 -alkoxy, fluorine, chlorine, bromine, trifluoromethyl, trifluoromethoxy or OH;
  • heteroaryl is pyridinyl or pyrimidinyl
  • aryl is phenyl or naphthyl
  • hetrocyclyl is morpholinyl, piperazinyl or piperidinyl;
  • A is A1 ;
  • R is unsubstituted or at least monosubstituted benzyl, phenylethyl-, phenylpropyl-, pyridinylmethyl-, pyridinylethyl- or pyridinylpropyl-,
  • substituents are selected from chlorine, bromine, fluorine, trifluoromethyl and carboxy;
  • Ar is unsubstituted or at least monosubstituted pyridin-4-yl, pyrimidin-4-yl or phenyl,
  • substituents are selected from methylamino-, ethylamino-, propylamino-, butylamino-, hydroxy, methoxy, ethoxy, methyl, ethyl, propyl,
  • the convertion with the amine (III) may be carried out using an inert solvent at 0 to 150 °C.
  • X is OH
  • the compounds of formula (I) may be obtained by acylation of the amine derivatives, either using an acid chloride to be added beforehand or by reaction in the presence of an activating agent.
  • the reaction may be carried out by forming an acid chloride according to any methods known to persons skilled in the art or more precisely by the action of oxalyl chloride in toluene, dichloromethane (R.D. MILLER, J. Org. Chem, 56, (4) 1453, (1991)) which, thus formed, will react with the amine (III) in the presence of a base such as pyridine, triethylamine, diisopropylethylamine; the reaction can start at 0°C and when the addition of the acid chloride is complete, the medium is kept stirred at room temperature (G. DAIDONE, Heterocycles, 43, (11), 2385-96, (1996)) or it is heated if necessary.
  • a base such as pyridine, triethylamine, diisopropylethylamine
  • the reaction may also be carried out in the presence of an activating agent of the carbodiimide type alone (DCC, EDAC) (M. C. DESAI, Tetrahedron Lett., 34, 7685, (1993)) or in the presence of hydroxybenzotriazole and dimethylaminopyridine (J. P. GAMET, Tetrahedron, 40, 1995, (1984), K. BARLOS, J. Org. Chem., 50, 696, (1985)) or according to well known methods of coupling in peptide chemistry (M. BODANSZKY, Principles of Peptide Synthesis; Springer-Verlag, New York, NY, pages 9-58, (1984)) or of forming the amide bond.
  • DCC carbodiimide type alone
  • EDAC EDAC
  • hydroxybenzotriazole and dimethylaminopyridine J. P. GAMET, Tetrahedron, 40, 1995, (1984), K. BARLOS, J. Org. Chem., 50
  • the derivatives of formula (II) are obtained by the method described in patent F.R 2481284 and by Y. Shojiro. Chem. Pharm. Bull; 19 (11) p 2354. It is necessary to protect the reactive functional groups.
  • the protecting groups are introduced according to any methods known to persons skilled in the art and in particular those described by T.W. GREENE, Protective groups in Organic Synthesis, J. Wiley-lnterscience Publication (1991).
  • For the phenols there will be preferably chosen more particularly a benzyl group introduced in the presence of an inorganic base such as sodium carbonate at the reflux temperature of acetone and of acetonitrile (A. R Mac Kenzie, Tetrahedron, 42, 3259, (1986)), which may then be removed by catalytic hydrogenation or more particularly using trifluoroacetic acid under reflux, described in patent W O 9727846.
  • the products of general formula (III) may be obtained commercially or by functionalization and protection of the reactive functional groups of commercially available products according to the methods described by Larock, Comprehensive Organic Transformations, VCH, New York, 1999.
  • the nitrile functional groups are reduced with hydrogen in the presence of catalysts, BH 3 or more precisely lithium aluminum hydride in solvents such as dioxane or THF (T.M. Koening, Tetrahedron Letters, 35, 1339, (1994)).
  • the phenol functional groups are protected with trimethylsilylethoxymethyl by reacting the starting compound with trimethylsilylethoxymethyl chloride in the presence of sodium hydride in a solvent such as dimethylformamide at room temperature (J. P. WHITTEN, J. Org.
  • the derivatives of formula (I) for which the protecting group is an ester can be saponified according to any methods known to persons skilled in the art and in particular by the action of sodium hydroxide on the reflux (L. Anzalone, J. Org. Chem., 50, 2128, (1985).
  • the derivatives of formula (I) may be obtained according to route a) by acylating the derivatives of formula (IV) either using an acid chloride, or according to the route b) by acylating the derivatives of formula (IV) or using an anhydride, or according to the route c) by the reaction of an acid in the presence of an activating agent.
  • the reaction is carried out in the presence of a base such as pyridine, triethylamine, diisopropylethylamine; the reaction may start at 0°C, and when the addition of the acid chloride is complete, the medium is kept stirred at room temperature (G. DAIDONE, Heterocycles, 43, (11), 2385-96, (1996) or it is heated if necessary.
  • a base such as pyridine, triethylamine, diisopropylethylamine
  • the reaction is carried out at the reflux temperature of an inert solvent such as xylene or tetrahydrofuran (F. ALBERICIO, Synth. Commun., 31 , (2), 225-32, (2001)) or dichloromethane, (G. PROCTER, Tetrahedron, 51 , (47), 12837-842, (1995)) or in the anhydride itself.
  • an inert solvent such as xylene or tetrahydrofuran (F. ALBERICIO, Synth. Commun., 31 , (2), 225-32, (2001)) or dichloromethane, (G. PROCTER, Tetrahedron, 51 , (47), 12837-842, (1995)) or in the anhydride itself.
  • the reaction is carried out in the presence of an activating agent of the carbodiimide type alone (DCC, EDAC) (M. C. DESAI, Tetrahedron Lett., 34, 7685, (1993)) or in the presence of hydroxybenzotriazole and dimethylaminopyridine (J. P. GAMET, Tetrahedron, 40, 1995, (1984), K. BARLOS, J. Org. Chem., 50, 696, (1985)) or according to well known methods of coupling in peptide chemistry (M. BODANSZKY, Principles of Peptide Synthesis; Springer- Verlag, New York, NY, pages 9-58, (1984)) or of forming the amide bond.
  • DCC carbodiimide type alone
  • EDAC EDAC
  • hydroxybenzotriazole and dimethylaminopyridine J. P. GAMET, Tetrahedron, 40, 1995, (1984), K. BARLOS, J. Org. Chem., 50, 6
  • compounds according to general formula (I) can be prepared by palladium catalyzed coupling according to a reaction of Suzuki (I. Parrot et al., Synthesis; 7; 1999; 1163-1168).
  • a compound of formula (IV), where Y1 is halogen, B(OH) or Sn(C C o-alkyl) and Y2 is H or a protecting group, is hereby converted with a compound of formula (V).
  • Z may be, for example, B(OH) 2 , B(C ⁇ -C 10 -alkyl) 2 , Sn(C ⁇ -C ⁇ 0 -alkyl) 3 , Zn(C ⁇ -C ⁇ o- alkyl) or halogen.
  • Y2 is a protecting group
  • said group is removed after the reaction of (IV) and (V) using methods known by a person skilled in the art. All protecting groups known by a person skilled in the art can be used as protecting groups, preferably trimethylsilylethoxymethyl-.
  • Pd(triphenylphosphin) 4 Pd-tetrakis-catalyst
  • the compounds of formula (I) are isolated and may be purified by known methods, for example by crystallization, chromatography or extraction.
  • the use of compounds according to the general formula (I) as pharmaceutical, where the compounds have the above-mentioned preferred, more preferred, even more preferred, even much more preferred, in particular preferred or exceptionally preferred meaning, are also subject of the present invention.
  • the compounds of general formula (I) are kinase inhibitors and can therefore be employed for the treatment of diseases, which may result from an abnormal activity of kinases.
  • abnormal kinase activity there may be mentioned, for example, that of PI3K, AkT, GSK-3 ⁇ and the like.
  • compounds according to the present invention can be used for the inhibition of the kinase GSK-3 ⁇ .
  • This effect is particularly relevant for the treatment of metabolic diseases such as type II diabetes or neurodegenerative diseases such as Alzheimer's disease.
  • compounds according to the general formula (I) have an inhibitory effect in respect of the phosphorylation of the tau-protein. This effect is particularly relevant for the treatment of neurodegenerative diseases such as Alzheimer's disease.
  • diseases which can be treated with the compounds according to the present invention, include: neurodegenerative diseases, strokes, cranial and spinal traumas and peripheral neuropathies, obesity, metabolic diseases, type II diabetes, essential hypertension, atherosclerotic cardiovascular diseases, polycystic ovary syndrome, syndrome X, immunodeficiency or cancer.
  • Neurodegenerative diseases are preferably: Alzheimer's disease, Parkinson's disease, frontoparietal dementia, corticobasal degeneration and Pick's disease.
  • Compounds according to the present invention are preferably employed for the treatment of metabolic diseases, in particular of type II diabetes.
  • the compounds according to the general formula (I) are preferably employed for the treatment of neurodegenerative diseases, in particular of Alzheimer's disease.
  • the item treatment also includes prophylaxis, therapy or curing of the above-mentioned diseases.
  • compositions for preparing one or more medicaments for prophylaxis and/or treatment of the before-mentioned diseases
  • pharmaceutical preparations comprising an effective dose of at least one compound of the formula (I) as well as pharmaceutical preparations comprising an effective dose of at least one compound of the formula (I) for prophylaxis and/or treatment of the before-mentioned diseases
  • the amount of a compound according to formula (I) which is required in order to attain the desired biological effect depends on a number of factors, for example the specific compound selected, the intended use, the type of administration and the clinical state of the patient.
  • the daily dose is in the range from 0.3 mg to 100 mg (typically from 3 mg to 50 mg) per day per kilogram of body weight, for example 3-10 mg/kg/day.
  • An intravenous dose can be, for example, in the range from 0.3 mg to 1.0 mg/kg and can be administered in a suitable manner as an infusion of 10 ng to 100 ng per kilogram per minute.
  • Suitable infusion solutions for these purposes may contain, for example, from 0.1 ng to 10 mg, typically from 1 ng to 10 mg per milliliter.
  • Individual doses may contain, for example, from 1 mg to 10 g of the active compound.
  • ampoules for injections can contain, for example, from 1 mg to 100 mg
  • orally administerable individual dose formulations such as, for example, tablets or capsules can contain, for example, from 1.0 to 1000 mg, typically from 10 to 600 mg.
  • the abovementioned masses relate to the mass of the free compound on which the salt is based.
  • the compound used for the prophylaxis or therapy of the abovementioned conditions may be the compounds according to formula (I) themselves, but they are preferably present in the form of a pharmaceutical composition together with an acceptable carrier.
  • the carrier must be naturally acceptable, in the sense that it is compatible with the other ingredients of said composition and is not harmful to the patient's health.
  • the carrier may be a solid or a liquid or both and is preferably formulated with the compound as an individual dose, for example as a tablet which may contain from 0.05% to 95% by weight of the active compound.
  • Further pharmaceutically active substances may also be present, including further compounds according to formula (I).
  • the pharmaceutical compositions of the invention may be prepared according to any of the known pharmaceutical methods which essentially comprise mixing the ingredients with pharmacologically acceptable carriers and/or excipients.
  • the pharmaceutical preparations can also contain additives.
  • additives can be employed, for example: fillers, binders, lubricants, wetting agents, stabilizers, emulsifiers, dispersants, preservatives, sweeteners, colorants, flavorings, aromatizers, thickeners, diluents, buffer substances, solvents, solubilizers, agents for achieving a depot effect, salts for altering the osmotic pressure, coating agents or antioxidants.
  • compositions of the invention may be in form of a pill, tablet, lozenge, coated tablet, granule, capsule, hard or soft gelatin capsule, aqueous solution, alcoholic solution, oily solution, syrup, emulsion suspension pastille suppository, solution for injection or infusion, ointment, tincture, cream, lotion, powder, spray, transdermal therapeutic systems, nasal spray, aerosol mixture, microcapsule, implant, rod or plaster.
  • compositions of the invention are those which are suitable for oral, rectal, topical, peroral (e.g. sublingual) and parenteral (e.g. subcutaneous, intramuscular, intradermal or intravenous) administration, although the most suitable manner of administration depends in each individual case on the nature and severity of the condition to be treated and on the nature of the compound according to formula (I) used in each case.
  • Sugar-coated formulations and sugar- coated delayed-release formulations are included within the scope of the invention.
  • Suitable pharmaceutical compounds for oral administration may be present in separate units as, for example, capsules, cachets, lozenges or tablets, which in each case contain a particular amount of the compound according to formula (I); as powders ( gelatin capsules or cachets) or granules; as solution or suspension in an aqueous or nonaqueous liquid; or as an oil-in-water or water-in-oil emulsion.
  • said compositions can be prepared according to any suitable pharmaceutical method which includes a step in which the active compound and the carrier (which may comprise one or more additional components) are contacted.
  • compositions are prepared by uniform and homogeneous mixing of the active compound with a liquid and/or finely dispersed solid carrier, after which the product is shaped, if necessary.
  • a tablet for example, may be prepared by pressing or shaping a powder or granules of the compound, where appropriate with one or more additional components. Pressed tablets can be prepared by tableting the compound in free-flowing form, for example a powder or granules, mixed, where appropriate, with a binder, lubricant, inert diluent and/or one or more surface active/dispersing agents in a suitable machine.
  • Shaped tablets can be prepared by shaping the pulverulent compound, moistened with an inert liquid diluent, in a suitable machine.
  • diluents can be used, for example, starch, cellulose, saccharose, lactose or silica.
  • the pharmaceutical compositions of the invention may also comprise substances other than diluents, for example one or more lubricants such as magnesium stearate or talc, a coloring, a coating (sugar-coated tablets) or a varnish.
  • compositions which are suitable for peroral (sublingual) administration include lozenges which contain a compound according to formula (I) with a flavoring, usually sucrose and gum arabic or tragacanth, and pastilles which comprise the compound in an inert base such as gelatin and glycerol or sucrose and gum arabic.
  • Suitable pharmaceutical compositions for parenteral administration preferably comprise sterile aqueous preparations of a compound according to formula (I) which are preferably isotonic with the blood of the intended recipient. These preparations are preferably administered intravenously, although they may also be administered subcutaneously, intramuscularly or intradermally as an injection. Said preparations may preferably be prepared by mixing the compound with water and rendering the obtained solution sterile and isotonic with the blood. Injectable compositions of the invention generally contain from 0.1 to 5% by weight of the active compound.
  • sterile compositions for parenteral administration may be preferably solutions which are aqueous or non aqueous, suspensions or emulsions.
  • solvent or vehicle there may be used water, propylene glycol, polyethylene glycol, vegetable oils, in particular olive oil, organic esters for injection, for example ethyl oleate or other suitable organic solvents.
  • These compositions may also contain adjuvants, in particular wetting, isotonizing, emulsifying, dispersing and stabilizing mediums.
  • the sterilization may be carried out in several ways, for example by an aseptic filtration, by incorporating sterilizing agents into the composition, by irradiation or by heating. They may also be prepared in the form of sterile solid compositions which may be dissolved at the time of use in sterile water or in any other sterile medium for injection.
  • Suitable pharmaceutical compositions for rectal administration are preferably present as individual dose suppositories. These may be prepared by mixing a compound according to formula (I) with one or more conventional solid carriers, for example cocoa butter, and shaping the resulting mixture.
  • Suitable pharmaceutical compositions for topical application to the skin are preferably present as ointment, cream, lotion, paste, spray, aerosol or oil.
  • Carriers which may be used are petroleum jelly, lanolin, polyethylene glycols, alcohols and combinations of two or more of these substances.
  • the active compound is present at a concentration of from 0.1 to 15%, for example from 0.5 to 2%, by weight of the composition.
  • Transdermal administration is also possible.
  • Suitable pharmaceutical compositions for transdermal administration may be present as individual patches which are suitable for long-term close contact with the epidermis of the patient.
  • patches suitably contain the active compound in an optionally buffered aqueous solution, dissolved and/or dispersed in an adhesive or dispersed in a polymer.
  • a suitable active compound concentration is from approx. 1 % to 35%, preferably approx. 3% to 15%.
  • a particular possibility is the release of the active compound by electro- transport or iontophoresis, as described, for example, in Pharmaceutical Research, 2(6): 318 (1986).
  • Tablets containing a dose of 50 mg of active product and having the following composition are prepared according to the usual technique:
  • a solution for injection containing 10 mg of active product and having the following composition is prepared:
  • Another subject of the present invention is the combination of compounds of the formula (I) with other pharmaceutically active substances not covered by formula
  • the compounds of the formula (I) are distinguished by beneficial actions on the metabolism of lipids, and they are particularly suitable for weight reduction and, after weight reduction, for maintaining a reduced weight in mammals and as anorectic agents.
  • the compounds are distinguished by their low toxicity and their few side effects.
  • the compounds may be employed alone or in combination with other weight- reducing or anorectic active compounds.
  • Further anorectic active compounds of this kind are mentioned, for example, in the Rote Liste, Chapter 01 under weight- reducing agents/appetite suppressants, and may also include those active compounds which increase the energy turnover of the organism and thus lead to weight reduction or else those which influence the general metabolism of said organism such that increased calorie intake does not cause an enlargement of the fat depots and a normal calorie intake causes a reduction in the fat depots of said organism.
  • the compounds are suitable for the prophylaxis and, in particular, for the treatment of problems of excess weight or obesity.
  • the compounds of formula (I) have a beneficial effect on the glucose metabolism, they particularly lower the blood-sugar level and can be used for treatment of type I and type II diabetes.
  • the compounds can therefore be used alone or in combination with other blood-sugar lowering active compounds (antidiabetics).
  • the compounds of the formula I may be administered in combination with one or more further pharmacologically active substances which may be selected, for example, from the group consisting of antidiabetics, antiadipose agents, blood-pressure-lowering active compounds, lipid reducers and active compounds for the treatment and/or prevention of complications caused by diabetes or associated with diabetes.
  • Suitable antidiabetics include insulins, amylin, GLP-1 and GLP-2 derivatives such as, for example, those disclosed by Novo Nordisk A/S in WO 98/08871 and also oral hypoglycemic active compounds.
  • Said oral hypoglycemic active compounds preferably include sulfonyl ureas, biguanidines, meglitinides, oxadiazolidinediones, thiazolidinediones, glucosidase inhibitors, glucagon receptor antagonists, GLP-1 agonists, potassium channel openers such as, for example, those disclosed by Novo Nordisk A/S in WO 97/26265 and WO 99/03861 , insulin sensitizers, activators of insulin receptor kinase, inhibitors of liver enzymes involved in the stimulation of gluconeogenesis and/or glycogenolysis, for example glycogen phosphorase inhibitors, modulators of glucose uptake and glucose elimination, lipid metabolism-modifying compounds such as antihyperlipidemic active compounds and antilipidemic active compounds, for example HMGCoA-reductase inhibitors, inhibitors of cholesterol transport/cholesterol uptake, inhibitors of the reabsorption of bile acid or inhibitors of microsomal
  • the present compounds are administered in combination with insulin.
  • the compounds of the invention are administered in combination with a sulfonylurea such as, for example, tolbutamide, glibenclamide, glimepiride, glipizide, gliquidone, glisoxepide, glibornuride or gliclazide.
  • a sulfonylurea such as, for example, tolbutamide, glibenclamide, glimepiride, glipizide, gliquidone, glisoxepide, glibornuride or gliclazide.
  • the compounds of the present invention are administered in combination with a biguanidine such as, for example, metformin.
  • the compounds of the present invention are administered in combination with a meglitinide such as, for example, repaglinide.
  • the compounds of the present invention are administered in combination with a thiazolidinedione such as, for example, troglitazone, ciglitazone, pioglitazone, rosiglitazone or the compounds disclosed by Dr. Reddy's Research Foundation in WO 97/41097, in particular 5-[[4-[(3,4-dihydro-3-methyl-4-oxo-2- quinazolinylmethoxy]phenyl]methyl]-2,4-thiazolidinedione.
  • the compounds of the present invention are administered in combination with an -glucosidase inhibitor such as, for example, miglitol or acarbose.
  • the compounds of the present invention are administered in combination with an active compound which acts on the ATP-dependent potassium channel of the beta cells, such as, for example, tolbutamide, glibenclamide, glimepiride, glipizide, gliclazide or repaglinide.
  • an active compound which acts on the ATP-dependent potassium channel of the beta cells such as, for example, tolbutamide, glibenclamide, glimepiride, glipizide, gliclazide or repaglinide.
  • the compounds of the present invention are administered in combination with an antihyperlipidemic active compound or an antilipidemic active compound such as, for example, cholestyramine, colestipol, clofibrate, fenofibrate, gemfibrozil, lovastatin, pravastatin, simvastatin, atorvastatin, cerivastatin, fluvastatin, probucol, ezetimibe or dextrothyroxine.
  • an antihyperlipidemic active compound or an antilipidemic active compound such as, for example, cholestyramine, colestipol, clofibrate, fenofibrate, gemfibrozil, lovastatin, pravastatin, simvastatin, atorvastatin, cerivastatin, fluvastatin, probucol, ezetimibe or dextrothyroxine.
  • the compounds of the present invention are administered in combination with more than one of the aforementioned compounds, for example in combination with a sulfonylurea and metformin, a sulfonylurea and acarbose, repaglinide and metformin, insulin and a sulfonylurea, insulin and metformin, insulin and troglitazone, insulin and lovastatin, etc.
  • the compounds of the invention may be administered in combination with one or more antiadipose agents or appetite-controlling active compounds.
  • Such active compounds may be selected from the group consisting of CART agonists, NPY antagonists, MC4 agonists, orexin antagonists, H3 agonists, TNF agonists, CRF agonists, CRF BP antagonists, urocortin agonists, ⁇ 3 agonists, MSH (melanocyte-stimulating hormone) agonists, CCK agonists, serotonin re- uptake inhibitors, mixed serotonin and noradrenalin reuptake inhibitors, 5HT modulators, MAO inhibitors, bombesin agonists, galanin antagonists, growth hormone, growth-hormone-releasing compounds, TRH agonists, uncoupling protein 2 or 3 modulators, leptin agonists, dopamine agonists (bromocriptine, doprexin), lipase/amylase inhibitors, cannabinoid receptor 1 antagonists, modulators of acylation-stimulating protein (ASP), PPAR modulators, RXR modulators
  • the antiadipose agent is leptin or modified leptin. In another embodiment, the antiadipose agent is dexamphetamine or amphetamine. In another embodiment, the antiadipose agent is fenfluramine or dexfenfluramine. In yet another embodiment, the antiadipose agent is sibutramine or the mono- and bis-demethylated active metabolite of sibutramine. In another embodiment, the antiadipose agent is orlistate. In another embodiment, the antiadipose agent is mazindol, diethylpropione or phentermine.
  • antihypertensive active compounds examples include betablockers such as alprenolol, atenol, timolol, pindolol, propanolol and metoprolol, ACE (angiotensin-converting enzyme) inhibitors such as, for example, benazepril, captopril, enalapril, fosinopril, lisinopril, quinapril and rampril, calcium channel blockers such as nifedipine, felodipine, nicardipine, isradipine, nimodipine, diltiazem and verapamil, and also alphablockers such as doxazosin, urapidil, prazosin and terazosin.
  • betablockers such as alprenolol, atenol, timolol, pindolol, propanolol and metoprolol
  • ACE angiotens
  • 0.14 g of 1 -hydroxybenzotriazole, 0.14 cm 3 of 2,4-dichlorobenzylamine and 0.14 cm 3 of triethylamine are added to 0.2 g of 3-oxo-6-phenyl-2,3-dihydro- pyridazine-4-carboxylic acid prepared as described by Y. Shojiro et al., Chem. Pharm. Bull; 19, (11), p. 2354, in 10 cm 3 of dichloromethane.
  • 0.2 g of 1-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride is then added. The mixture is stirred for 48 hours at 19°C. 10 cm 3 of distilled water are added.
  • the organic phase is washed again with 3 times 10 cm 3 of aqueous normal hydrochloric acid solution and then with 10 cm 3 of saturated aqueous sodium chloride solution.
  • the organic phase is then dried over magnesium sulfate.
  • the mixture is filtered through a sinter funnel and then evaporated under reduced pressure (2 kPa; 45°C).
  • the residue is taken up with 10 cm 3 of diisopropyl ether.
  • the insoluble material is filtered off through a sinter funnel and then rinsed again with 10 cm 3 of diisopropyl ether.
  • reaction mixture is then poured onto a solution of 10 cm 3 of dichloromethane containing 0.19 cm 3 of triethylamine and 0.21 cm 3 of 2,4-dichlorobenzylamine.
  • the reaction medium is stirred for 12 hours at 19°C and then filtered through a sinter funnel, rinsed with 10 cm 3 of dichloromethane, 10 cm 3 of distilled water and with 10 cm 3 of aqueous normal hydrochloric acid solution.
  • the organic phase is dried over magnesium sulfate, filtered through a sinter funnel and then evaporated under reduced pressure (2 kPa; 45°C).
  • the solid residue is triturated in 20 cm 3 of pentane, suction-filtered through a sinter funnel and oven- dried under reduced pressure (10 kPa; 20°C). 23.1 g of 4-benzyloxyacetophenone are obtained in the form of a white solid melting at 99°C.
  • the product obtained is triturated in 150 cm 3 of ethanol, filtered through a sinter funnel, washed with twice 50 cm 3 of ethanol and 50 cm 3 of isopropyl ether to give, after drying under reduced pressure (2 kPa; 55°C), 5.6 g of diethyl hydroxy[2-(4-benzyloxyphenyl)-2-oxoethyl]malonate melting at 80°C.
  • N-(2,4-dichlorobenzyl)-3-oxo-6-[4-(hydroxy)phenyl]-2,3-dihydropyri- dazine-4-carboxamide is obtained in the form of a yellow solid melting at a temperature above 260°C.
  • Oxalyl chloride is added to a solution a 8.73 g of 6-chloro-3-oxo-2,3- dihydro-pyridazine-4-carboxylic acid and 1 ml DMF in 250 ml THF at 5- 10°C and the mixture is stirred at room temperature for 2 h. Afterwards, it is evaporated to dryness, the residue dissolved in 450 ml THF and 13.8 g potassium carbonate and a solution of 7.2 g 4-chloro-benzyl amide in THF are added. The solvent is distilled off after 2 h of stirring at room temperature, the residue suspended in 100 ml water and a pH of 6.4 is adjusted. The obtained precipitate is sucked off, suspended again in 50 ml
  • the cortex sections having a thickness of 300 ⁇ m are prepared from 8-10- week old male OFA rats (Iffa-Credo), sacrificed by decapitation. They are incubated in 5 ml of DMEM medium containing pyruvate and glucose 4.5 g/l at 37°C for 40 min. The sections are then washed twice with the medium, distributed into microtubes (50 ⁇ l in 500 ⁇ l of medium with or without test compounds), and incubated at 37°C, with stirring. Two hours later, the experiment is stopped by centrifugation.
  • the sections are washed, sonicated and centrifuged at 18300 g, for 15 min at 4°C.
  • concentration of proteins in supernatant is determined by a commercial assay (BCA Protein Assay, Pierce) based on the Lowry method.
  • the samples denatured beforehand for 10 min at 70°C, are separated on a 4-12% Bis-Tris vertical gel in the presence of MOPS-SDS buffer and electrotransferred onto nitrocellulose membrane.
  • Immunolabeling is performed with their monoclonal antibody AD2 which specifically recognizes the phosphorylated epitopes Ser396/404 of the Tau protein.
  • the immunoactive proteins are visualized by adding a second antibody directed against the mouse ⁇ y a s and coupled to peroxidase and a chemiluminescent substrate.
  • the autoradiograms obtained are finally quantified using the 'GeneTools' software from Syngene (GeneGnome, Ozyme) in order to determine an IC 50 .
  • the compounds of formula (I) have a very advantageous activity and in particular some compounds have an IC 50 of less than 100 ⁇ M.
  • GSK- ⁇ activity is measured using human recombinant GSK-3B and a primed (pre-phosphorylated) substrate peptide (derived from glycogen synthase and containing the phosphorylation sites 3a, b, and c) on basis of the AlphaScreen technology in 384-well plate format (small volume plate, white, GREINER).
  • a primed (pre-phosphorylated) substrate peptide derived from glycogen synthase and containing the phosphorylation sites 3a, b, and c
  • 384-well plate format small volume plate, white, GREINER
  • phospho-glycogen synthase peptide 34 nM in kinase buffer (20 mM Hepes, pH 7,4, 10 mM MgCI, 200 mM EDTA, 1 mM DTT, 0,1 mg/ml BSA, 10 ⁇ M ATP) are incubated at room temperature for 60 min.

Abstract

The present invention relates to novel pyridazinone derivatives of the general formula (I) wherein A is A1 or A2; R is unsubstituted or at least monosubstituted C1-C10-alkyl, aryl, aryl (C1-C10-alkyl)-, heteroaryl, heteroaryl-(C1-C10-­alkyl)-, heterocyclyl, heterocyclyl-(C1-C10-alkyl)-, C3-C10-cycloalkyl, polycycloalkyl, C2-C10 alkenyl or C2-C10-alkinyl and Ar is unsubstituted or at least monosubstituted aryl or heteroaryl.

Description

PYRIDAZINONE DERIVATIVES AS GS -3BETA INHIBITORS
The present invention relates to compounds according to the general formula (I), with the definitions of the substituents A and Ar given below in the text, as well as their physiologically acceptable salts, methods for producing these compounds and their use as pharmaceuticals.
Figure imgf000002_0001
These compounds are kinase inhibitors, in particular inhibitors of the kinase GSK- 3β (glycogen synthase kinase-3β).
It is known from literature that in the case of metabolic diseases such as diabetes or neurodegenerative diseases such as Alzheimer's disease, there is a connection between the therapy of said diseases and the inhibition of GSK-3β or the phosphorylation of the tau-protein (S.E. Nikoulina. Diabetes 51 , 2190-2198, 2002; Henrikson. Am. J. Physiol. 284, E892-900, 2003). Many compounds or pharmaceuticals, respectively, are already known to be employed for the treatment of said diseases, which compounds interfere at different places of the biochemical processes causing the respective disease. However, there are no compounds known until now, which effect inhibition of GSK-3β.
Pyridazinone derivatives are well known pharmaceuticals, but it has not been reported so far that pyridazinone derivatives can be employed for the inhibition of GSK-3β or tau-phosporylation, respectively. Pyridazinone derivatives described in literature differ from those of the present invention due to a different substitution pattern and (partially) different indications.
WO 03/059891 discloses pyridazinone derivatives that are useful for treating diseases and conditions caused or exacerbated by unregulated p38 MAP Kinase activity and/or TNF activity. The compounds described therein can be used, for example, for the treatment of inflammatory conditions, diabetes, Alzheimer's disease or cancer. They differ from the compounds of the present invention in the substitution of the pyridazinone cycle, since the nitrogen at position 2 of the cycle is mostly substituted with alky, aryl or heteroaryl and at position 4 of the cycle there is no amido group defined as substituent (equals substituent A of the compounds of the present invention).
The documents EP-A 075 436, US 4,734,415 and US 4,353,905 describe pyridazinone derivatives as antihypertensive agents and as agents which increase cardiac contractibility. These pyridazinone derivatives have a phenyl residue at position 6 of the pyridazinone cycle, said phenyl residue is additionally substituted with a heterocycle containing at least one nitrogen atom. Whereas the pyridazinone derivatives described in the documents EP-A 075 436 and US 4,353,905 do not have a substituent at position 4 of the pyridazinone cycle, those disclosed in US 4,734,415 may have an amido group substituted with lower alkyl at this position. Compounds as such, explicitly disclosed by US 4,743,415, are not a subject of the present invention.
Thus, there exists a strong need for compounds having an inhibitory effect for GSK-3β and/or the phosphorylation of the tau-protein. The object of the present invention is to provide compounds showing this ability.
This object is attained by pyridazinone derivatives according to the below- mentioned formula (I)
Figure imgf000003_0001
wherein A represents A1 or A2
Figure imgf000004_0001
R is unsubstituted or at least monosubstituted Ci-Cio-alkyl, aryl, aryl-(Cι-Cι0- alkyl)-, heteroaryl, heteroaryl-(Cι-Cιo-alkyl)-, heterocyclyl, heterocyclyl-(Cr C10-alkyl)-, C3-Cι0-cycloalkyl, polycycloalkyl, C2-Cι0-alkenyl or C2-Cι0- alkinyl,
where the substituents are selected from halogen, -CN, C Cι0-alkyl, -N02, - OR1 , -C(0)OR1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -SR1 , - S(0)R1 , -S02R1 , -NHS02R1 , -S02NR1 R2, -C(S)NR1 R2, -NHC(S)R1 ,
-O-SO2RI , -SO2-O-RI , oxo, -C(0)R1 , -C(NH)NH2, heterocyclyl, C3-C10- cycloalkyl, aryl-(CrC6-alkyl)-, aryl, heteroaryl, trifluoromethyl, trifluoromethylsulfanyl and trifluoromethoxy,
and aryl, heterocyclyl and heteroaryl may in turn be at least monosubstituted with Ci-Cβ-alkyl, Ci-Cβ-alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
Ar is unsubstituted or at least monosubstituted aryl or heteroaryl;
where the substituents are selected from halogen, -CN, NO2, CrCι0-alkyl, -OR1 , -C(0)OR1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -NHC(S)R1 , -C(S)NR1 R2, -SR1 , -S(0)R1 , -S02R1 , -NHS02R1 , -S02NR1 R2, -0-S02R1 , -SO2-O-RI , aryl, heteroaryl, ary CrC-e-alkyl)-, formyl, trifluoromethyl and trifluoromethoxy,
and aryl and heteroaryl may in turn be at least monosubstituted with Cι-C6- alkyl, CrCβ-alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
R1 and R2 are independently from each other
hydrogen;
unsubstituted or at least monosubstituted Cι-Cι0-alkyl, C3-Cι0-cycloalkyl, aryl, aryl-(CrCιo-alkyl)-, C2-C10-alkenyl, C2-Cι0-alkinyl, heterocyclyl, heterocyclyl-(Ci-Cio-alkyl)- or heteroaryl, where the substituents are selected from halogen, Cι-C6-alkyl, Ci-Ce-alkoxy, CN, N02 , NH2, (Cι-C6- alkyl)amino-, d d-Ce-alky amino-, OH, COOH, -COO-(Cι-C6-alkyl), -CONH2, formyl, trifluoromethyl and trifluoromethoxy;
heteroaryl is a 5 to 10-membered, aromatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O und S;
aryl is phenyl, indanyl, indenyl or naphthyl;
heterocyclyl is a 5 to 10-membered, aliphatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O and S;
with the proviso that A is not -C(0)NH(Cι-Cβ-alkyl), in case Ar is phenyl which is at least monosubstituted with heterocyclyl or heteroaryl containing nitrogen.
If in the compounds of formula (I), groups, fragments, residues or substituents such as, for example, aryl, heteroaryl, alkyl etc., may be present several times, they all independently from each other have the meanings indicated and may hence, in each individual case, be identical with or different from each other. The following comments apply to (for example) aryl as well as to any other residue independently from its classification as aryl group, -substituent, -fragment or - residue. One example is the di(CrCi0-alkyl)amino group in which the alkyl substitutents may be identical or different (for instance 2 x ethyl or 1 x propyl and 1 x heptyl).
If in the above-mentioned definitions of compounds according to formula (I) a substituent, for example aryl, may be unsubstituted or at least mono-substituted with a group of further substituents, for example, Cι-C6-alkyl, Cι-C6-alkoxy, halogen etc., it applies in such cases, where there is a poly-substitution of aryl, that the selection from the group of further substituents is independently from each other. Thus, all combinations of further substituents are comprised in the case of, for example, a double-substitution of aryl. Therefore, aryl may be substituted twice with ethyl, aryl may be mono-substituted with methyl or ethoxy, respectively, aryl may be mono-substituted with ethyl or fluoro, respectively, aryl may be substituted twice with methoxy, etc.. Alkyl, alkenyl and alkynyl residues may be linear or branched. This also applies when they are part of other groups, for example in alkoxy groups (C Cι0-alkyl-O-), alkoxycarbonyl groups or amino groups, or when they are substituted.
Examples for alkyl groups are: methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl. This comprises both the n-isomers of these residues and isopropyl, isobutyl, isopentyl, sec-butyl, tert-butyl, neopentyl, 3,3-dimethylbutyl etc.. Furthermore, unless stated otherwise, the term alkyl here also includes unsubstituted alkyl residues as well as alkyl residues which are substituted by one or more, for example one, two, three or four, identical or different residues, for example aryl, heteroaryl, alkoxy or halogen. The substituents may be present in any desired position of the alkyl group.
Examples for cycloalkyl residues are: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. All cycloalkyl groups may be unsubstituted or optionally substituted by one or more further residues, as exemplified above in the case of the alkyl groups.
Examples for alkenyl and alkynyl groups are the vinyl residue, the 1-propenyl residue, the 2-propenyl residue (allyl residue), the 2-butenyl residue, the 2-methyl- 2-propenyl residue, the 3-methyl-2-butenyl residue, the ethynyl residue, the 2- propynyl residue (propargyl residue), the 2-butynyl residue or the 3-butynyl residue. The term alkenyl here also expressly includes cycloalkenyl residues and cycloalkenyl-alkyl-residues (alkyl substituted by cycloalkenyl) containing at least three carbon atoms. Examples for cycloalkenyl residues are cyclopentenyl, cyclohexenyl, cycloheptenyl and cyclooctenyl.
The alkenyl residues may have 1 to 3 conjugated or unconjugated double bonds in a straight or branched chain; the same applies to alkynyl residues in respect of triple bonds. The alkenyl and alkinyl residues may be unsubstituted or optionally substituted by one or more further residues, as exemplified above in the case of the alkyl groups.
Examples for polycycloalkyl residues are: adamantyl, quinuclidinyl, bornanyl, norbornanyl, bornenyl and norbornenyl. If not stated otherwise, the above-mentioned aryl, heteroaryl and heterocyclic residues may be unsubstituted or may carry one or more, for example one, two, three or four of the substituents indicated in the above definition, which substituents may be in any desired position. In monosubstituted phenyl residues, for example, the substituent may be in the 2-position, the 3-position or the 4- position, in disubstituted phenyl residues the substituents may be in 2,3-position, 2,4-position, 2,5-position, 2,6-position, 3,4-position or 3,5-position. In trisubstituted phenyl residues the substituents may be in 2,3,4-position, 2,3,5-position, 2,3,6- position, 2,4,5-position, 2,4,6-position or 3,4,5-position. In fourfold substituted phenyl residues, the substituents may be in the 2,3,4,5-position, the 2,3,4,6- position, or the 2, 3,5,6-position.
The above definitions as well as the following definitions relating to monovalent residues equally apply to the divalent residues phenylene, naphthylene and heteroarylene. Those divalent residues (fragments) may be attached to the adjacent groups by any ring carbon atom. In the case of a phenylene residue, these may be in 1 , 2-position (ortho-phenylene), 1 , 3-position (meta-phenylene) or 1 ,4-position (para-phenylene). In the case of 5-membered ring aromatics containing one heteroatom such as, for example, thiophene or furan, the two free bonds may be in 2,3-position, 2,4-position, 2,5-position or 3,4-position. A divalent residue derived from pyridine may be a 2,3-, 2,4-, 2,5-, 2,6-, 3,4- or 3,5-pyridinediyl residue. In the case of unsymmetrical divalent residues the present invention includes all positional isomers, i. e., in the case of a 2,3-pyridinediyl residue, for example, it includes the compound in which the one adjacent group is present in the 2-position and the other adjacent group is present in the 3-position as well as the compound in which the one adjacent group is present in the 3-position and the other adjacent group is present in the 2-position.
Unless stated otherwise, heteroaryl residues, heteroarylene residues, heterocyclyl residues, heterocyclylen residues and rings which are formed by two groups bonded to a nitrogen are preferably derived from completely saturated, partially saturated or completely unsaturated heterocycles (i.e. heterocycloalkanes, heterocycloalkenes, heteroaromatics), which contain one, two, three or four heteroatoms, which may be identical or different; more preferably they are derived from heterocycles which contain one, two, or three, in particular one or two, heteroatoms, which may be identical or different. Unless stated otherwise, the heterocycles may be monocyclic or polycyclic, for example monocyclic, bicyclic or tricyclic. Preferably they are monocyclic or bicyclic. The rings preferably are 5- membered rings, 6-membered rings or 7-membered rings. Examples of monocyclic and bicyclic heterocyclic systems from which residues occuring in the compounds of the formula (I) may be derived, are pyrrole, furan, thiophene, imidazole, pyrazole, 1 ,2,3-triazole, 1 ,2,4-triazole, 1 ,3-dioxole, 1 ,3-oxazole (= oxazole), 1 ,2-oxazole (= isoxazole), 1 ,3-thiazole (= thiazole), 1 ,2-thiazole (= isothiazole), tetrazole, pyridine, pyridazine, pyrimidine, pyrazine, pyran, thiopyran, 1 ,4-dioxine, 1 ,2-oxazine, 1 ,3-oxazine, 1 ,4-oxazine, 1 ,2-thiazine, 1 ,3-thiazine, 1 ,4- thiazine, 1 ,2,3-triazine, 1 ,2,4-triazine, 1 ,3,5-triazine, 1 ,2,4,5-tetrazine, azepine, 1 ,2- diazepine, 1 ,3-diazepine, 1 ,4-diazepine, 1 ,3-oxazepine, 1 ,3-thiazepine, indole, benzothiophene, benzofuran, benzothiazole, benzimidazole, benzodioxol, quinoline, isoquinoline, quinazoline, quinoxaline, phthalazine, thienothiophenes, 1 ,8-naphthyridine and other naphthyridines, pteridin, or phenothiazine, each of them in saturated form (perhydro form) or in partially unsaturated form (for example in the dihydro form or the tetrahydro form) or in maximally unsaturated form, in case the respective forms are known and stable. The term "aryl" and the term "heteroaryl" as used herein comprise bicyclic residues in which both cycles are aromatic as well as bicyclic residues in which only one cycle is aromatic. Suitable aliphatic heterocycles include, for example, the saturated heterocycles pyrrolidine, piperidine, piperazine, imidazolidine, pyrazolidine, isothiazolidine, thiazolidine, isoxazolidine, oxazolidine, tetrahydrofuran, dioxolane, 2-oxo-azepane, morpholine and thiomorpholine as well as the partially unsaturated heterocycles isochromamyl, chromamyl, 1 ,2,3,4-tetrahydroisochinolyl and 1 ,2,3,4- tetrahydrochinolyl. The degree of saturation of heterocyclic groups is indicated in their individual definitions.
Substituents which may be derived from these heterocycles may be attached via any suitable carbon atom. Residues derived from nitrogen heterocycles may carry a hydrogen atom or a substituent on a ring nitrogen atom, and examples include pyrrole, imidazole, pyrrolidine, morpholine, piperazine residues, etc. Those nitrogen heterocyclic residues may also be attached via a ring nitrogen atom, in particular if the respective heterocyclic residue is bonded to a carbon atom. For example, a thienyl residue may be present as 2-thienyl or 3-thienyl, a piperidinyl residue as 1 -piperidinyl (= piperidino), 2-piperidinyl, 3-piperidinyl or 4-piperidinyl. Suitable nitrogen heterocycles may also be present as N-oxides or as quarternary salts containing a counterion which is derived from a physiologically acceptable acid. Pyridyl residues, for example, may be present as pyridine-N-oxides. Arylalkyl means an alkyl residue, which in turn is substituted by an aryl residue. Heteroarylalkyl means an alkyl residue, which in turn is substituted by a heteroaryl residue. Heterocyclylalkyl means an alkyl residue, which in turn is substituted by a heterocyclyl residue. For the definitions and possible substitutions of alkyl, heteroaryl, heterocyclyl and aryl it is referred to the above-mentioned definitions.
Halogen is fluorine, chlorine, bromine or iod, preferably fluorine, chlorine or bromine, most preferably fluorine or chlorine.
The present invention includes all stereoisomeric forms of the compounds of the formula (I). Centers of asymmetry that are present in the compounds of formula (I) all independently of one another have S configuration or R configuration. The invention includes all possible enantiomers and diastereomers and mixtures of two or more stereoisomers, for example mixtures of enantiomers and/or diastereomers, in all ratios. Thus, compounds according to the present invention which may exist as enantiomers may be present in enantiomerically pure form, both as levorotatory and as dextrorotatory antipodes, in the form of racemates and in the form of mixtures of the two enantiomers in all ratios. In the case of a cis/trans isomerism the invention includes both the cis form and the trans form as well as mixtures of these forms in all ratios. All these forms are an object of the present invention. The preparation of individual stereoisomers may be carried out, if desired, by separation of a mixture by customary methods, for example by chromatography or crystallization, by the use of stereochemically uniform starting materials for the synthesis or by stereoselective synthesis. Optionally, a derivatization may be carried out before a separation of stereoisomers. The separation of a mixture of stereoisomers may be carried out at the stage of the compounds of the formula (I) or at the stage of an intermediate during the synthesis. The present invention also includes all tautomeric forms of the compounds of formula (I), in particular keto-enol tautomehsm, i.e. the respective compounds may be present either in their keto form or in their enol form or in mixtures thereof in all ratios.
In case the compounds according to formula (I) contain one or more acidic or basic groups, the invention also comprises their corresponding physiologically or toxicologically acceptable salts. Physiologically acceptable salts are particularly suitable for medical applications, due to their greater solubility in water compared with the starting or base compounds. Said salts must have a physiologically acceptable anion or cation. Suitable physiologically acceptable acid addition salts of the compounds of the invention are salts of inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, metaphosphoric acid, nitric acid, sulfonic acid and sulfuric acid and also of organic acids such as, for example, acetic acid, theophyllinacetic acid, methylene-bis-b-oxynaphthoic acid, benzenesulfonic acid, benzoic acid, citric acid, ethanesulfonic acid, salicylic acid, fumaric acid, gluconic acid, glycolic acid, isethionic acid, lactic acid, lactobionic acid, maleic acid, malic acid, methane- sulfonic acid, succinic acid, p-toluenesulfonic acid, tartaric acid and trifluoroacetic acid. Suitable pharmaceutically acceptable basic salts are ammonium salts, alkali metal salts (such as sodium salts and potassium salts) and alkaline earth metal salts (such as magnesium salts and calcium salts).
Salts having a pharmaceutically unacceptable anion are likewise included within the scope of the present invention as useful intermediates for preparing or purifying pharmaceutically acceptable salts and/or for use in nontherapeutic applications, for example in-vitro applications.
If the compounds of the formula (I) simultaneously contain acidic and basic groups in the molecule, the invention also includes, in addition to the salt forms mentioned, inner salts or betaines (zwitterions).
The respective salts according to the formula (I) may be obtained by customary methods which are known to the person skilled in the art like, for example by contacting these with an organic or inorganic acid or base in a solvent or dispersant, or by anion exchange or cation exchange with other salts.
The present invention furthermore includes all solvates of compounds of the formula (I), for example hydrates or adducts with alcohols, active metabolites of the compounds of the formula (I), and also derivatives, which contain physiologically tolerable and cleavable groups, for example esters or amides.
The term "physiologically functional derivative" used herein relates to any physiologically acceptable derivative of an inventive compound of the formula I, for example an ester which on administration to a mammal, for example humans, is capable of forming (directly or indirectly) a compound of the formula I or an active metabolite thereof.
The physiologically functional derivatives also include prodrugs of the compounds of the invention. Such prodrugs may be metabolized in vivo to a compound of the invention. These prodrugs may or may not be active themselves and are also object of the present invention.
The compounds of the invention may also be present in various polymorphous forms, for example as amorphous and crystalline polymorphous forms. All polymorphous forms of the compounds of the invention are included within the scope of the invention and are another aspect of the invention.
Compounds of formula (I) are preferred, wherein
A is A1 ;
R is unsubstituted or at least monosubstituted CrCio-alkyl, aryl, aryl-(Cι-Cιo- alkyl)-, heteroaryl, heteroaryl-(Ci-Cio-alkyl)-, heterocyclyl, heterocyclyl- (C-i- Cio-alkyl)-, C3-Cιo-cycloalkyl, polycycloalkyl, C2-Cιo-alkenyl or C-2-C10- alkinyl,
where the substituents are selected from halogen, -CN, d-Cio-alkyl, -NO2, -OR1 , -C(0)OR1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -SR1 , -S(0)R1 , -SO2RI , -NHSO2RI . -S02NR1 R2, -C(S)NR1 R2, -NHC(S)R1 ,
-0-S02R1 , -SO2-O-RI , oxo, -C(0)R1 , -C(NH)NH2, heterocyclyl, C3-C10- cycloalkyl, aryl-(Cι-C6-alkyl)-, aryl, heteroaryl, trifluoromethyl, trifluoromethylsulfanyl and trifluoromethoxy,
and aryl, heterocyclyl and heteroaryl may in turn be at least monosubstituted with C-i-Cβ-alkyl, CrC6-alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
R1 and R2 independently from each other
hydrogen; unsubstituted or at least monosubstituted CrCio-alkyl, C3-Cι0-cycloalkyl, aryl, aryl-(CrC10-alkyl)-, C2-Cι0-alkenyl, C2-Cι0-alkinyl, heterocyclyl, heterocyclyl-(CrCιo-alkyl)- or heteroaryl, where the substituents are selected from halogen, Cι-C6-alkyl, CrC6-alkoxy, CN, N02 , NH2, (C C6- alkyl) amino-, d d-Ce-alkylJamino-, OH, COOH, -COO-(CrC6-alkyl), -CONH2, formyl, trifluoromethyl and trifluoromethoxy;
heteroaryl is a 5 to 10-membered, aromatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O and S;
aryl is phenyl, indanyl, indenyl or naphthyl;
heterocyclyl is a 5 to 10-membered, aliphatic, mono- Oder bicyclic heterocycle containing one or more heteroatoms selected from N, O and S;
In another embodiment, compounds of formula (I) are preferred, wherein
R is unsubstituted or at least monosubstituted Ci-Cio-alkyl, aryl, aryl-(Cι-Cιo- alkyl)-, heterocyclyl, heterocyclyl-(CrCi0-alkyl)-, C3-Cι.0-cycloalkyl, heteroaryl or heteroaryl-(CrCιo-alkyl)-,
where the substituents are selected from halogen, -CN, Cι-Cι0-Alkyl, -NO2, -OR1 , -C(0)OR1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -SR1 , -S(0)R1 , -SO2RI , -NHSO2RI , -S02NR1 R2, -C(S)NR1 R2, -NHC(S)R1 , -O-SO2RI , -SO2-O-RI , oxo, -C(0)R1 , -C(NH)NH2, heterocyclyl, C3-C10- cycloalkyl, aryl-(d~C6-alkyl)-, aryl, heteroaryl, trifluoromethyl, trifluoromethylsulfanyl and trifluoromethoxy,
and aryl, heterocyclyl and heteroaryl may in turn be at least monosubstituted with CrCβ-alkyl, d-C-β-alkoxy, halogen, trifluoromethyl, trifluoroethoxy or OH;
R1 and R2 are independently from each other
hydrogen; unsubstituted or at least monosubstituted d-Cio-alkyl, C3-Cι0-cycloalkyl, aryl, aryl-(d-Cio-alkyl)-, C2-do-alkenyl, C2-C10-alkinyl, heterocyclyl, heterocyclyl-(Cι-Cιo-alkyl)- or heteroaryl, where the substituents are selected from halogen, Cι-C6-alkyl, d-C6-alkoxy, CN, N02 , NH2, (Cι-C6- alkyl)ami no-, di(d-C6-alkyl)amino-, OH, COOH, -COO-(d-C6-alkyl), -CONH2, formyl, trifluoromethyl and trifluoromethoxy;
heteroaryl is a 5 to 10-membered, aromatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O and S;
aryl is phenyl, indanyl, indenyl or naphthyl;
heterocyclyl is a 5 to 10-membered, aliphatic, mono- Oder bicyclic heterocycle, containing one or more heteroatoms selected from N, 0 and S;
In another embodiment, compounds of formula (I) are preferred, wherein
Ar is unsubstituted or at least monosubstituted phenyl, pyridinyl, pyrimidinyl, pyrazolyl, thiophenyl, isoxaloyl, benzo[b]thiophenyl, benzodioxolyl or thiazolo[3,2-b][1 ,2,4]-tiazolyl,
where the substituents are selected from halogen, -CN, N02, CrCio-alkyl, -OR1 , -C(0)0R1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -NHC(S)R1 , -C(S)NR1 R2, -SR1 , -S(0)R1 , -S02R1 , -NHS02R1 , -SO2NRI R2, -O-SO2RI , -SO2-O-RI , aryl, heteroaryl, aryl-(d-C6-alkyl)-, formyl, trifluoromethyl and trifluoromethoxy,
and aryl and heteroaryl may in turn be at least monosubstituted with d-C6- alkyl, Cι-C6-alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
R1 and R2 are independently from each other
hydrogen;
unsubstituted or at least monosubstituted C Cio-alkyl, C3-Cι0-cycloalkyl, aryl, aryl-(d-C10-alkyl)-, C2-Cι0-alkenyl, C2-Cι0-alkinyl, heterocyclyl, heterocyclyl-(d-do-alkyl)- or heteroaryl, where the substituents are selected from halogen, Cι-C6-alkyl, C C6-alkoxy, CN, N02 , NH2, (C C6- alkyl) amino-, di(d-C6-alkyl)amino-, OH, COOH, -COO-(d-C6-alkyl), -CONH2, formyl, trifluoromethyl and trifluoromethoxy;
heteroaryl is a 5 to 10-membered aromatic, mono- or bicyclic heterocycle, containing one or more heteroatoms selected from N, O and S;
aryl is phenyl, indanyl, indenyl or naphthyl;
heterocyclyl is a 5 to 10-membered aliphatic, mono- oder bicyclic heterocycle, containing one or more heteroatoms selected from N, O and S;
Compounds of formula (I) are more preferred, wherein
A is A1 ;
R is unsubstituted or at least monosubstituted CrCio-alkyl, aryl, aryl-(Cι~Cιo- alkyl)-, heterocyclyl, heterocyclyl-(Cι-Cιo-alkyl)-, C3-Cι0-cycloalkyl, heteroaryl or heteroaryl-(Ci-Cio-alkyl)-,
where the substituents are selected from halogen, CrCio-alkyl, -OR1 , -C(0)OR1 , -NR1 R2, -C(0)NR1 R2, -SR1 , -S02R1 , -S02NR1 R2, oxo, -C(0)R1 , -C(NH)NH2, heterocyclyl, C3-C10-cycloalkyl, aryl-(d-C6-alkyl)-, aryl, trifluoromethyl and trifluoromethoxy,
and aryl, heterocyclyl and heteroaryl may in turn be at least monosubtituted with d-C-6-alkyl, d-C-β-alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
Ar is unsubstituted or at least monosubstituted phenyl, pyridinyl, pyrimidinyl, pyrazolyl, thiophenyl, isoxazolyl, benzo[b]thiophenyl, benzodioxolyl or thiazolo[3,2-b][1 ,2,4]-tiazolyl,
where the substituents are selected from halogen, Ci-Cio-alkyl, -OR1 , -C(0)OR1 , -NR1 R2, -C(0)NR1 R2, aryl, heteroaryl, aryl-(d-C6-alkyl)-, trifluoromethyl and Trifluoromethoxy, and aryl and heteroaryl may in turn be at least monosubsituted with Cι-C6- alkyl, d-C6-alkoxy, halogen, trifluoromethyl or OH;
R1 und R2 are independently from each other
hydrogen;
unsubstituted or at least monosubstituted d-Cio-alkyl, C3-Cιo~cycloalkyl, aryl, aryl-(Cι-Cιo-alkyl)-, heterocyclyl, heterocyclyl-(Cι-Cιo-alkyl)- or heteroaryl, where the substituents are selected from halogen, d-C6-alkyl, d-Ce-alkoxy, NH2, (d-C6-alkyl)amino-, di(d-C6-alkyl)amino-, OH, trifluoromethyl and trifluoromethoxy;
heteroaryl is a 5 to 10-membered, aromatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O und S; heteroaryl is preferably imidazolyl, thiophenyl, furanyl, isoxazolyl, pyridinyl, pyrimidinyl, 1 ,2,3,4- tetrahydrochinolinyl, benzoimidazolyl, indolyl or benzodioxolyl;
aryl is phenyl, indanyl, indenyl or naphthyl; aryl is preferably phenyl or naphthyl.
heterocyclyl is a 5 to 10-membered, aliphatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O und S; heterocyclyl is preferably 2-oxo-azepanyl, tetrahydrofuranyl, 1 ,3-dioxolanyl, morpholinyl, piperazinyl or piperidinyl;
Compounds of formula (I) are even more preferred, wherein
A is A1 ;
R is unsubstituted or at least monosubstituted d-Cio-alkyl, aryl, aryl-(Cι-Cιo- alkyl)-, heterocyclyl, heterocyclyl-(d-C10-alkyl)-, C3-Cιo-cycloalkyl, heteroaryl or heteroaryl-(Cι-Cιo-alkyl)-,
where the substituents are selected from halogen, C Cio-alkyl, -OR1 , -C(0)OR1 , -NR1 R2, -C(0)NR1 R2, -SR1 , -S02R1 , -S02NR1 R2, oxo,
-C(0)R1 , -C(NH)NH2, heterocyclyl, C3-Cι0-cycloalkyl, aryl-(C C6-alkyl)-, aryl, trifluoromethyl and trifluoromethoxy, and aryl, heterocyclyl and heteroaryl may in turn be at least monosubsituted with Ci-Ce-alkyl, Cι-C6-alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
Ar is unsubstituted or at least monosubstituted phenyl, pyridinyl or pyrimidinyl,
where the substituents are selected from halogen, Ci-Cio-alkyl, -OR1 , -C(0)OR1 , -NR1 R2, -C(0)NR1 R2, aryl, heteroaryl, aryl-(d-C6-alkyl)-, trifluoromethyl and trifluoromethoxy,
and aryl and heteroaryl may in turn be at least monosubsituted with Ci-Ce- alkyl, Ci-Cβ-alkoxy, halogen, trifluoromethyl or OH;
R1 und R2 are independently from each other
hydrogen;
unsubstituted or at least monosubstituted Ci-Cio-alkyl, C3-do-cycloalkyl, aryl, aryl-(Cι-Cιo-alkyl)-, heterocyclyl, heterocyclyl-(Ci-Cio-alkyl)- or heteroaryl, where the substituents are selected from halogen, Ci-Ce-alkyl, Ci-Ce-alkoxy, NH2, (Cι-C6-alkyl)amino-, di(d-C6-alkyl)amino-, OH, trifluoromethyl and trifluoromethoxy;
heteroaryl is a 5 to 10-membered, aromatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O und S; heteroaryl is preferably imidazolyl, thiophenyl, furanyl, isoxazolyl, pyridinyl, pyrimidinyl, 1 ,2,3,4- tetrahydrochinolinyl, benzoimidazolyl, indolyl or benzodioxolyl;
aryl is phenyl, indanyl, indenyl or naphthyl; aryl is preferably phenyl or naphthyl.
heterocyclyl is a 5 to 10-membered, aliphatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O und S; heterocyclyl is preferably 2-oxo-azepanyl, tetrahydrofuranyl, 1 ,3-dioxolanyl, morpholinyl, piperazinyl or piperidinyl;
Compounds of formula (I) are even much more preferred, wherein A is A1 ;
R is unsubstituted or at least monosubstituted aryl-(Cι-C-6-alkyl)- or heteroaryl- (Ci-Ce-alkyl)-,
where the substituents are selected from halogen, Cι-C6-alkyl, -OH, -O-aryl, Ci-Ce-alkoxy, -0-(Cι-C6-alkylen)-N(Cι-C6-alkyl)2, -C(0)OH, -C(0)0-(Cι-C6- alkyl), -NH2, -N(Cι-C6-alkyl)2, -NH(Cι-C6-alkyl), -NH(d-C10-cycloalkyl), -C(0)NH2, -C(0)NH-heteroaryl, -C(0)NH-(Ci-C6-alkyl), -S02(d-Ce-alkyl),
-S02NH2, -C(0)-heterocyclyl, -C(NH)NH2, heterocyclyl, aryl-(d-C6-alkyl)-, aryl, trifluoromethyl, and trifluoromethoxy,
and aryl, aeterocyclyl and heteroaryl may in turn be at least monosubstituted with d-C3-alkyl, Cι-C3-alkoxy, fluorine, chlorine, bromine, trifluoromethyl, trifluoromethoxy or OH;
heteroaryl is imidazolyl, thiophenyl, furanyl, isoxazolyl, pyridinyl, pyrimidinyl, benzoimidazolyl, indolyl or benzodioxolyl;
aryl is phenyl or naphthyl;
hetrocyclyl is morpholinyl, piperazinyl or piperidinyl;
In another embodiment, compounds of formula (I) are even much more preferred, wherein
A is A1 ;
Ar is unsubstituted or at least monosubstituted phenyl, pyridin-4-yl or pyrimidin-4-yl,
where the substituents are selected from halogen, Ci-Ce-alkyl, -OH, Ci-Ce- alkoxy, -C(0)OH, -C(0)0-(Cι-C6-alkyl), -NH2, -N(C -C6-alkyl)2, -NH(d-C6- alkyl), -NH(Ci-Cio-cycloalkyl), -NH(heterocyclyl-(d-C6-alkyl-)), -NH(aryl-
(Cι-C6-alkyl-)), -C(0)NH2, -C(0)NH-(d-C6-alkyl), aryl, and heteroaryl, and aryl, heterocyclyl and heteroaryl may in turn be at least monosubstituted Cι-C3-Alkyl, d-C3-alkoxy, fluorine, chlorine, bromine, trifluoromethyl, trifluoromethoxy or OH;
heteroaryl is pyridinyl or pyrimidinyl;
aryl is phenyl or naphthyl;
hetrocyclyl is morpholinyl, piperazinyl or piperidinyl;
Compounds of formula (I) are particularly preferred, wherein
A is A1 ;
R is unsubstituted or at least monosubstituted benzyl, phenylethyl-, phenylpropyl-, pyridinylmethyl-, pyridinylethyl- or pyridinylpropyl-,
where the substituents are selected from chlorine, bromine, fluorine, trifluoromethyl and carboxy;
Ar is unsubstituted or at least monosubstituted pyridin-4-yl, pyrimidin-4-yl or phenyl,
where the substituents are selected from methylamino-, ethylamino-, propylamino-, butylamino-, hydroxy, methoxy, ethoxy, methyl, ethyl, propyl,
(phenylethyl)amino-, benzylamino- and (morpholinylethyl)amino-;
Compounds of formula (I) are exceptionally preferred, which are selected from the group consisting of
6-(2-butylamino-pyrimidin-4-yl)-3-oxo-2,3-dihydro-pyridazine-4-car- boxylic acid (3-pyridin- 3-yl-propyl)-amide,
6-(4-hydroxy-3-methoxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid (3-pyridin-3-yl-propyl)-amide,
6-(4-hydroxy-3,5-dimethyl-phenyl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid (3-pyridin-3-yl-propyl)-amide, 6-(4-hydroxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4-carboxylic acid (3- pyridin-3-yl-propyl)-amide,
6-(2-ethylamino-pyrimidin-4-yl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid 4-chloro- benzylamide,
6-(3-chloro-4-hydroxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4-carboxylic acid 4-chloro-benzylamide,
6-(4-hydroxy-3,5-dimethyl-phenyl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid 4-chloro-benzylamide,
4-({[6-(4-hydroxy-3,5-dimethyl-phenyl)-3-oxo-2,3-dihydro-pyridazine-4- carbonyl]-amino}- methyl)-benzoic acid,
4-({[6-(4-hydroxy-3-methoxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4- carbonylj-amino}- methyl)-benzoic acid,
6-(2-butylamino-pyrimidin-4-yl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid (pyridin-3- ylmethyl)-amide,
6-(3-fluoro-4-hydroxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4-carboxylic acid 4-chloro-benzylamide,
6-(4-hydroxy-3-methyl-phenyl)-3-oxo-2,3-dihydro-pyridazine-4-carboxylic acid 4-chloro-benzylamide,
6-[2-(2-morpholin-4-yl-ethylamino)-pyrimidin-4-yl]-3-oxo-2,3-dihydro- pyridazine-4- carboxylic acid 4-chloro-benzylamide,
6-(4-hydroxy-3-methoxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid 4-chloro-benzylamide,
6-(2-methylamino-pyrimidin-4-yl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid 4-chloro-benzylamide, R-3-oxo-6-[2-(1-phenyl-ethylamino)-pyrimidin-4-yl]-2,3-dihydro- pyridazine-4-carboxylic acid (3-phenyl-propyl)-amide,
6-(4-hydroxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4-carboxylic acid 4- chloro-benzylamide,
3-oxo-6-pyridin-4-yl-N-[4-(trifluoromethyl)benzyl]-2,3-dihydropyridazine- 4-carboxamide,
3-oxo-6-pyridin-4-yl-2,3-dihydro-pyridazine-4-carboxylic acid 4-bromo- benzylamide,
3-oxo-6-pyridin-4-yl-N-(pyridin-3-ylmethyl)-2,3-dihydropyridazine-4- carboxamide,
N-(2,4-dichlorobenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4- carboxamide,
3-oxo-6-pyridin-4-yl-2,3-dihydro-pyridazine-4-carboxylic acid 4-chloro-2- fluoro-benzylamide, and
N-(4-chlorobenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4- carboxamide;
It is explicitly indicated once more that also in case of the preferred, more preferred, even more preferred, even much more preferred, particularly preferred and exceptionally preferred compounds according to formula (I) the above- mentioned explanations also apply in respect of the salts, stereoisomers, prodrugs, N-oxides etc.; in particular, the respective physiologically acceptable salts are included.
The derivatives of formula (I) for which A = A1= CONHR may be obtained from compounds according to formula (II), where X is a functional group, preferably - OH, Cι-Cιo-alkoxy, chloro or -O-C(O)-(Cι-Cι0-alkyl). The convertion with the amine (III) may be carried out using an inert solvent at 0 to 150 °C. In case X is OH, the compounds of formula (I) may be obtained by acylation of the amine derivatives, either using an acid chloride to be added beforehand or by reaction in the presence of an activating agent.
Figure imgf000021_0001
The reaction may be carried out by forming an acid chloride according to any methods known to persons skilled in the art or more precisely by the action of oxalyl chloride in toluene, dichloromethane (R.D. MILLER, J. Org. Chem, 56, (4) 1453, (1991)) which, thus formed, will react with the amine (III) in the presence of a base such as pyridine, triethylamine, diisopropylethylamine; the reaction can start at 0°C and when the addition of the acid chloride is complete, the medium is kept stirred at room temperature (G. DAIDONE, Heterocycles, 43, (11), 2385-96, (1996)) or it is heated if necessary.
The reaction may also be carried out in the presence of an activating agent of the carbodiimide type alone (DCC, EDAC) (M. C. DESAI, Tetrahedron Lett., 34, 7685, (1993)) or in the presence of hydroxybenzotriazole and dimethylaminopyridine (J. P. GAMET, Tetrahedron, 40, 1995, (1984), K. BARLOS, J. Org. Chem., 50, 696, (1985)) or according to well known methods of coupling in peptide chemistry (M. BODANSZKY, Principles of Peptide Synthesis; Springer-Verlag, New York, NY, pages 9-58, (1984)) or of forming the amide bond.
The derivatives of formula (II) are obtained by the method described in patent F.R 2481284 and by Y. Shojiro. Chem. Pharm. Bull; 19 (11) p 2354. It is necessary to protect the reactive functional groups. The protecting groups are introduced according to any methods known to persons skilled in the art and in particular those described by T.W. GREENE, Protective groups in Organic Synthesis, J. Wiley-lnterscience Publication (1991). For the phenols, there will be preferably chosen more particularly a benzyl group introduced in the presence of an inorganic base such as sodium carbonate at the reflux temperature of acetone and of acetonitrile (A. R Mac Kenzie, Tetrahedron, 42, 3259, (1986)), which may then be removed by catalytic hydrogenation or more particularly using trifluoroacetic acid under reflux, described in patent W O 9727846.
The products of general formula (III) may be obtained commercially or by functionalization and protection of the reactive functional groups of commercially available products according to the methods described by Larock, Comprehensive Organic Transformations, VCH, New York, 1999. The nitrile functional groups are reduced with hydrogen in the presence of catalysts, BH3 or more precisely lithium aluminum hydride in solvents such as dioxane or THF (T.M. Koening, Tetrahedron Letters, 35, 1339, (1994)). The phenol functional groups are protected with trimethylsilylethoxymethyl by reacting the starting compound with trimethylsilylethoxymethyl chloride in the presence of sodium hydride in a solvent such as dimethylformamide at room temperature (J. P. WHITTEN, J. Org. Chem., 51, 1891 , (1986); M. P. EDWARDS, Tetrahedron, 42, 3723, (1986)). The deprotection is carried out according to methods known to persons skilled in the art and described by T.W. GREENE, Protective groups in Organic Synthesis, J. Wiley-lnterscience Publication (1991).
The derivatives of formula (I) for which the protecting group is trimethylsilylethoxymethyl can be deprotected by reaction with tetrabutylammonium fluoride under reflux in solvents such as tetrahydrofuran, dioxane. (J. P. WHITTEN, J. Org. Chem., 51, 1891 , (1986); B. H. LIPSHUTZ, Tetrahedron Lett., 4095, (1986)).
The derivatives of formula (I) for which the protecting group is an ester can be saponified according to any methods known to persons skilled in the art and in particular by the action of sodium hydroxide on the reflux (L. Anzalone, J. Org. Chem., 50, 2128, (1985).
For the derivatives in formula (I) for which A = A2 = NHCOR, it is necessary to subject the derivatives of formula (II) to a rearrangement according to the methods described by Larock, Comprehensive Organic Transformations, VCH, New York, 1999 or more particularly by B. Singh, HETEROCYCLES, 31, (12), 2163, (1990).
Figure imgf000023_0001
The derivatives of formula (I) may be obtained according to route a) by acylating the derivatives of formula (IV) either using an acid chloride, or according to the route b) by acylating the derivatives of formula (IV) or using an anhydride, or according to the route c) by the reaction of an acid in the presence of an activating agent.
ent
Figure imgf000023_0003
Figure imgf000023_0002
By the route (a) the reaction is carried out in the presence of a base such as pyridine, triethylamine, diisopropylethylamine; the reaction may start at 0°C, and when the addition of the acid chloride is complete, the medium is kept stirred at room temperature (G. DAIDONE, Heterocycles, 43, (11), 2385-96, (1996) or it is heated if necessary.
By the route (b) the reaction is carried out at the reflux temperature of an inert solvent such as xylene or tetrahydrofuran (F. ALBERICIO, Synth. Commun., 31 , (2), 225-32, (2001)) or dichloromethane, (G. PROCTER, Tetrahedron, 51 , (47), 12837-842, (1995)) or in the anhydride itself.
By the route (c) the reaction is carried out in the presence of an activating agent of the carbodiimide type alone (DCC, EDAC) (M. C. DESAI, Tetrahedron Lett., 34, 7685, (1993)) or in the presence of hydroxybenzotriazole and dimethylaminopyridine (J. P. GAMET, Tetrahedron, 40, 1995, (1984), K. BARLOS, J. Org. Chem., 50, 696, (1985)) or according to well known methods of coupling in peptide chemistry (M. BODANSZKY, Principles of Peptide Synthesis; Springer- Verlag, New York, NY, pages 9-58, (1984)) or of forming the amide bond. Furthermore, compounds according to general formula (I) can be prepared by palladium catalyzed coupling according to a reaction of Suzuki (I. Parrot et al., Synthesis; 7; 1999; 1163-1168). A compound of formula (IV), where Y1 is halogen, B(OH) or Sn(C C o-alkyl) and Y2 is H or a protecting group, is hereby converted with a compound of formula (V).
Figure imgf000024_0001
Z may be, for example, B(OH)2, B(Cι-C10-alkyl)2, Sn(Cι-Cι0-alkyl)3, Zn(Cι-Cιo- alkyl) or halogen. In case Y2 is a protecting group, said group is removed after the reaction of (IV) and (V) using methods known by a person skilled in the art. All protecting groups known by a person skilled in the art can be used as protecting groups, preferably trimethylsilylethoxymethyl-. For performing the palladium catalyzed coupling all palladium complexes known by a person skilled in the art can be employed, preferably Pd(triphenylphosphin)4 (Pd-tetrakis-catalyst) is employed, which is preferably obtained in situ from palladium acetate.
The compounds of formula (I) are isolated and may be purified by known methods, for example by crystallization, chromatography or extraction.
Subject of the present invention is also the use of compounds according to the general formula (I) as pharmaceuticals or medicaments, respectively. With respect to the definition of the substituents A and Ar (as well as all further substituents defined by the before-mentioned substituents) the same explanations as laid out above in the context with the compounds as such apply.
The use of compounds according to the general formula (I) as pharmaceutical, where the compounds have the above-mentioned preferred, more preferred, even more preferred, even much more preferred, in particular preferred or exceptionally preferred meaning, are also subject of the present invention. The compounds of general formula (I) are kinase inhibitors and can therefore be employed for the treatment of diseases, which may result from an abnormal activity of kinases. As abnormal kinase activity, there may be mentioned, for example, that of PI3K, AkT, GSK-3β and the like.
In particular, compounds according to the present invention can be used for the inhibition of the kinase GSK-3β. This effect is particularly relevant for the treatment of metabolic diseases such as type II diabetes or neurodegenerative diseases such as Alzheimer's disease.
Furthermore, compounds according to the general formula (I) have an inhibitory effect in respect of the phosphorylation of the tau-protein. This effect is particularly relevant for the treatment of neurodegenerative diseases such as Alzheimer's disease.
Examples of diseases, which can be treated with the compounds according to the present invention, include: neurodegenerative diseases, strokes, cranial and spinal traumas and peripheral neuropathies, obesity, metabolic diseases, type II diabetes, essential hypertension, atherosclerotic cardiovascular diseases, polycystic ovary syndrome, syndrome X, immunodeficiency or cancer. Neurodegenerative diseases are preferably: Alzheimer's disease, Parkinson's disease, frontoparietal dementia, corticobasal degeneration and Pick's disease.
Compounds according to the present invention are preferably employed for the treatment of metabolic diseases, in particular of type II diabetes.
In another embodiment of the present invention, the compounds according to the general formula (I) are preferably employed for the treatment of neurodegenerative diseases, in particular of Alzheimer's disease.
In the above-mentioned explanation the item treatment also includes prophylaxis, therapy or curing of the above-mentioned diseases.
All references to "compound(s) according to formula (I)" refer hereinbelow to a compound/compounds of the formula (I) as described above and also to their salts, solvates and physiologically functional derivatives as described herein. The compounds of the formula (I) can be administered to animals, preferably to mammals, and in particular to humans. The compounds of the formula (I) can be administered as pharmaceuticals by themselves, in mixtures with one another or in mixtures with other pharmaceuticals or in the form of pharmaceutical preparations. Further subjects of the present invention therefore also are the use of the compounds of the formula (I) for preparing one or more medicaments for prophylaxis and/or treatment of the before-mentioned diseases, pharmaceutical preparations (or pharmaceutical compositions) comprising an effective dose of at least one compound of the formula (I) as well as pharmaceutical preparations comprising an effective dose of at least one compound of the formula (I) for prophylaxis and/or treatment of the before-mentioned diseases
The amount of a compound according to formula (I) which is required in order to attain the desired biological effect depends on a number of factors, for example the specific compound selected, the intended use, the type of administration and the clinical state of the patient. In general, the daily dose is in the range from 0.3 mg to 100 mg (typically from 3 mg to 50 mg) per day per kilogram of body weight, for example 3-10 mg/kg/day. An intravenous dose can be, for example, in the range from 0.3 mg to 1.0 mg/kg and can be administered in a suitable manner as an infusion of 10 ng to 100 ng per kilogram per minute. Suitable infusion solutions for these purposes may contain, for example, from 0.1 ng to 10 mg, typically from 1 ng to 10 mg per milliliter. Individual doses may contain, for example, from 1 mg to 10 g of the active compound. Thus, ampoules for injections can contain, for example, from 1 mg to 100 mg, and orally administerable individual dose formulations such as, for example, tablets or capsules can contain, for example, from 1.0 to 1000 mg, typically from 10 to 600 mg. In the case of pharmaceutically acceptable salts, the abovementioned masses relate to the mass of the free compound on which the salt is based. The compound used for the prophylaxis or therapy of the abovementioned conditions may be the compounds according to formula (I) themselves, but they are preferably present in the form of a pharmaceutical composition together with an acceptable carrier. The carrier must be naturally acceptable, in the sense that it is compatible with the other ingredients of said composition and is not harmful to the patient's health. The carrier may be a solid or a liquid or both and is preferably formulated with the compound as an individual dose, for example as a tablet which may contain from 0.05% to 95% by weight of the active compound. Further pharmaceutically active substances may also be present, including further compounds according to formula (I). The pharmaceutical compositions of the invention may be prepared according to any of the known pharmaceutical methods which essentially comprise mixing the ingredients with pharmacologically acceptable carriers and/or excipients.
Besides at least one compound according to formula (I) as well as one or more carriers, the pharmaceutical preparations can also contain additives. As additives can be employed, for example: fillers, binders, lubricants, wetting agents, stabilizers, emulsifiers, dispersants, preservatives, sweeteners, colorants, flavorings, aromatizers, thickeners, diluents, buffer substances, solvents, solubilizers, agents for achieving a depot effect, salts for altering the osmotic pressure, coating agents or antioxidants.
The pharmaceutical compositions of the invention may be in form of a pill, tablet, lozenge, coated tablet, granule, capsule, hard or soft gelatin capsule, aqueous solution, alcoholic solution, oily solution, syrup, emulsion suspension pastille suppository, solution for injection or infusion, ointment, tincture, cream, lotion, powder, spray, transdermal therapeutic systems, nasal spray, aerosol mixture, microcapsule, implant, rod or plaster.
Pharmaceutical compositions of the invention are those which are suitable for oral, rectal, topical, peroral (e.g. sublingual) and parenteral (e.g. subcutaneous, intramuscular, intradermal or intravenous) administration, although the most suitable manner of administration depends in each individual case on the nature and severity of the condition to be treated and on the nature of the compound according to formula (I) used in each case. Sugar-coated formulations and sugar- coated delayed-release formulations, too, are included within the scope of the invention. Preference is given to acid-resistant and enteric formulations. Suitable enteric coatings include cellulose acetate phthalate, polyvinyl acetate phthalate, hydroxypropyl-methylcellulose phthalate and anionic polymers of methacrylic acid and methyl methacrylate.
Suitable pharmaceutical compounds for oral administration may be present in separate units as, for example, capsules, cachets, lozenges or tablets, which in each case contain a particular amount of the compound according to formula (I); as powders ( gelatin capsules or cachets) or granules; as solution or suspension in an aqueous or nonaqueous liquid; or as an oil-in-water or water-in-oil emulsion. As already mentioned, said compositions can be prepared according to any suitable pharmaceutical method which includes a step in which the active compound and the carrier (which may comprise one or more additional components) are contacted. In general, the compositions are prepared by uniform and homogeneous mixing of the active compound with a liquid and/or finely dispersed solid carrier, after which the product is shaped, if necessary. Thus a tablet, for example, may be prepared by pressing or shaping a powder or granules of the compound, where appropriate with one or more additional components. Pressed tablets can be prepared by tableting the compound in free-flowing form, for example a powder or granules, mixed, where appropriate, with a binder, lubricant, inert diluent and/or one or more surface active/dispersing agents in a suitable machine. Shaped tablets can be prepared by shaping the pulverulent compound, moistened with an inert liquid diluent, in a suitable machine. As diluents can be used, for example, starch, cellulose, saccharose, lactose or silica. The pharmaceutical compositions of the invention may also comprise substances other than diluents, for example one or more lubricants such as magnesium stearate or talc, a coloring, a coating (sugar-coated tablets) or a varnish.
Pharmaceutical compositions which are suitable for peroral (sublingual) administration include lozenges which contain a compound according to formula (I) with a flavoring, usually sucrose and gum arabic or tragacanth, and pastilles which comprise the compound in an inert base such as gelatin and glycerol or sucrose and gum arabic.
Suitable pharmaceutical compositions for parenteral administration preferably comprise sterile aqueous preparations of a compound according to formula (I) which are preferably isotonic with the blood of the intended recipient. These preparations are preferably administered intravenously, although they may also be administered subcutaneously, intramuscularly or intradermally as an injection. Said preparations may preferably be prepared by mixing the compound with water and rendering the obtained solution sterile and isotonic with the blood. Injectable compositions of the invention generally contain from 0.1 to 5% by weight of the active compound.
These sterile compositions for parenteral administration may be preferably solutions which are aqueous or non aqueous, suspensions or emulsions. As solvent or vehicle, there may be used water, propylene glycol, polyethylene glycol, vegetable oils, in particular olive oil, organic esters for injection, for example ethyl oleate or other suitable organic solvents. These compositions may also contain adjuvants, in particular wetting, isotonizing, emulsifying, dispersing and stabilizing mediums. The sterilization may be carried out in several ways, for example by an aseptic filtration, by incorporating sterilizing agents into the composition, by irradiation or by heating. They may also be prepared in the form of sterile solid compositions which may be dissolved at the time of use in sterile water or in any other sterile medium for injection.
Suitable pharmaceutical compositions for rectal administration are preferably present as individual dose suppositories. These may be prepared by mixing a compound according to formula (I) with one or more conventional solid carriers, for example cocoa butter, and shaping the resulting mixture.
Suitable pharmaceutical compositions for topical application to the skin are preferably present as ointment, cream, lotion, paste, spray, aerosol or oil. Carriers which may be used are petroleum jelly, lanolin, polyethylene glycols, alcohols and combinations of two or more of these substances. In general, the active compound is present at a concentration of from 0.1 to 15%, for example from 0.5 to 2%, by weight of the composition.
Transdermal administration is also possible. Suitable pharmaceutical compositions for transdermal administration may be present as individual patches which are suitable for long-term close contact with the epidermis of the patient. Such patches suitably contain the active compound in an optionally buffered aqueous solution, dissolved and/or dispersed in an adhesive or dispersed in a polymer. A suitable active compound concentration is from approx. 1 % to 35%, preferably approx. 3% to 15%. A particular possibility is the release of the active compound by electro- transport or iontophoresis, as described, for example, in Pharmaceutical Research, 2(6): 318 (1986).
The following examples illustrate compositions according to the invention:
EXAMPLE A Gelatin capsules containing a dose of 50 mg of active product and having the following composition are prepared according to the usual technique:
- Compound of formula (I) 50 mg - Cellulose 18 mg
- Lactose 55 mg
- Colloidal silica 1 mg
- Sodium carboxymethylstarch 10 mg
- Talc 10 mg - Magnesium stearate 1 mg
EXAMPLE B
Tablets containing a dose of 50 mg of active product and having the following composition are prepared according to the usual technique:
- Compound of formula (I) 50 mg
- Lactose 104 mg
- Cellulose 40 mg - Polyvidone 10 mg
- Sodium carboxymethylstarch 22 mg
- Talc 10 mg
- Magnesium stearate 2 mg
- Colloidal silica 2 mg - Mixture of hydroxymethylcellulose, glycerin, titanium oxide
(72-3.5-24.5) qs 1 finished film-coated tablet of 245 mg
EXAMPLE C
A solution for injection containing 10 mg of active product and having the following composition is prepared:
- Compound of formula (I) 10 mg
- Benzoic acid 80 mg - Benzyl alcohol 0.06 ml
- Sodium benzoate 80 mg
- Ethanol at 95 % 0.4 ml - Sodium hydroxide 24 mg
- Propylene glycol 1.6 ml
- Water qs 4 ml
Another subject of the present invention is the combination of compounds of the formula (I) with other pharmaceutically active substances not covered by formula
(0-
The compounds of the formula (I) are distinguished by beneficial actions on the metabolism of lipids, and they are particularly suitable for weight reduction and, after weight reduction, for maintaining a reduced weight in mammals and as anorectic agents. The compounds are distinguished by their low toxicity and their few side effects.
The compounds may be employed alone or in combination with other weight- reducing or anorectic active compounds. Further anorectic active compounds of this kind are mentioned, for example, in the Rote Liste, Chapter 01 under weight- reducing agents/appetite suppressants, and may also include those active compounds which increase the energy turnover of the organism and thus lead to weight reduction or else those which influence the general metabolism of said organism such that increased calorie intake does not cause an enlargement of the fat depots and a normal calorie intake causes a reduction in the fat depots of said organism. The compounds are suitable for the prophylaxis and, in particular, for the treatment of problems of excess weight or obesity.
The compounds of formula (I) have a beneficial effect on the glucose metabolism, they particularly lower the blood-sugar level and can be used for treatment of type I and type II diabetes. The compounds can therefore be used alone or in combination with other blood-sugar lowering active compounds (antidiabetics). In a further aspect of the invention, the compounds of the formula I may be administered in combination with one or more further pharmacologically active substances which may be selected, for example, from the group consisting of antidiabetics, antiadipose agents, blood-pressure-lowering active compounds, lipid reducers and active compounds for the treatment and/or prevention of complications caused by diabetes or associated with diabetes. Suitable antidiabetics include insulins, amylin, GLP-1 and GLP-2 derivatives such as, for example, those disclosed by Novo Nordisk A/S in WO 98/08871 and also oral hypoglycemic active compounds.
Said oral hypoglycemic active compounds preferably include sulfonyl ureas, biguanidines, meglitinides, oxadiazolidinediones, thiazolidinediones, glucosidase inhibitors, glucagon receptor antagonists, GLP-1 agonists, potassium channel openers such as, for example, those disclosed by Novo Nordisk A/S in WO 97/26265 and WO 99/03861 , insulin sensitizers, activators of insulin receptor kinase, inhibitors of liver enzymes involved in the stimulation of gluconeogenesis and/or glycogenolysis, for example glycogen phosphorase inhibitors, modulators of glucose uptake and glucose elimination, lipid metabolism-modifying compounds such as antihyperlipidemic active compounds and antilipidemic active compounds, for example HMGCoA-reductase inhibitors, inhibitors of cholesterol transport/cholesterol uptake, inhibitors of the reabsorption of bile acid or inhibitors of microsomal triglyceride transfer protein (MTP), compounds which reduce food intake, PPAR and RXR agonists and active compounds which act on the ATP- dependent potassium channel of beta cells.
In one embodiment of the present invention, the present compounds are administered in combination with insulin.
In another embodiment, the compounds of the invention are administered in combination with a sulfonylurea such as, for example, tolbutamide, glibenclamide, glimepiride, glipizide, gliquidone, glisoxepide, glibornuride or gliclazide. In another embodiment, the compounds of the present invention are administered in combination with a biguanidine such as, for example, metformin. In another embodiment, the compounds of the present invention are administered in combination with a meglitinide such as, for example, repaglinide. In yet another embodiment, the compounds of the present invention are administered in combination with a thiazolidinedione such as, for example, troglitazone, ciglitazone, pioglitazone, rosiglitazone or the compounds disclosed by Dr. Reddy's Research Foundation in WO 97/41097, in particular 5-[[4-[(3,4-dihydro-3-methyl-4-oxo-2- quinazolinylmethoxy]phenyl]methyl]-2,4-thiazolidinedione. In another embodiment, the compounds of the present invention are administered in combination with an -glucosidase inhibitor such as, for example, miglitol or acarbose.
In another embodiment, the compounds of the present invention are administered in combination with an active compound which acts on the ATP-dependent potassium channel of the beta cells, such as, for example, tolbutamide, glibenclamide, glimepiride, glipizide, gliclazide or repaglinide. In yet another embodiment, the compounds of the present invention are administered in combination with an antihyperlipidemic active compound or an antilipidemic active compound such as, for example, cholestyramine, colestipol, clofibrate, fenofibrate, gemfibrozil, lovastatin, pravastatin, simvastatin, atorvastatin, cerivastatin, fluvastatin, probucol, ezetimibe or dextrothyroxine.
In another embodiment, the compounds of the present invention are administered in combination with more than one of the aforementioned compounds, for example in combination with a sulfonylurea and metformin, a sulfonylurea and acarbose, repaglinide and metformin, insulin and a sulfonylurea, insulin and metformin, insulin and troglitazone, insulin and lovastatin, etc.
Furthermore, the compounds of the invention may be administered in combination with one or more antiadipose agents or appetite-controlling active compounds.
Such active compounds may be selected from the group consisting of CART agonists, NPY antagonists, MC4 agonists, orexin antagonists, H3 agonists, TNF agonists, CRF agonists, CRF BP antagonists, urocortin agonists, β3 agonists, MSH (melanocyte-stimulating hormone) agonists, CCK agonists, serotonin re- uptake inhibitors, mixed serotonin and noradrenalin reuptake inhibitors, 5HT modulators, MAO inhibitors, bombesin agonists, galanin antagonists, growth hormone, growth-hormone-releasing compounds, TRH agonists, uncoupling protein 2 or 3 modulators, leptin agonists, dopamine agonists (bromocriptine, doprexin), lipase/amylase inhibitors, cannabinoid receptor 1 antagonists, modulators of acylation-stimulating protein (ASP), PPAR modulators, RXR modulators, hCNTF mimetics or TR-β agonists.
In one embodiment of the invention, the antiadipose agent is leptin or modified leptin. In another embodiment, the antiadipose agent is dexamphetamine or amphetamine. In another embodiment, the antiadipose agent is fenfluramine or dexfenfluramine. In yet another embodiment, the antiadipose agent is sibutramine or the mono- and bis-demethylated active metabolite of sibutramine. In another embodiment, the antiadipose agent is orlistate. In another embodiment, the antiadipose agent is mazindol, diethylpropione or phentermine.
Furthermore, the compounds of the present invention may be administered in combination with one or more antihypertensive active compounds. Examples of antihypertensive active compounds are betablockers such as alprenolol, atenol, timolol, pindolol, propanolol and metoprolol, ACE (angiotensin-converting enzyme) inhibitors such as, for example, benazepril, captopril, enalapril, fosinopril, lisinopril, quinapril and rampril, calcium channel blockers such as nifedipine, felodipine, nicardipine, isradipine, nimodipine, diltiazem and verapamil, and also alphablockers such as doxazosin, urapidil, prazosin and terazosin. Furthermore, reference may be made to Remington: The Science and Practice of Pharmacy, 19th edition, Gennaro, editor, Mack Publishing Co., Easton, PA, 1995.
It is self-evident that every suitable combination of the compounds of the invention with one or more of the aforementioned compounds and optionally one or more other pharmacologically active substances is to be regarded as covered by the scope of protection of the present invention.
The following examples illustrate the invention without limitation.
Example 1
N-(2,4-dichlorobenzyl)-3-oxo-6-phenyl-4-yl-2-3-dihydropyridazine-4- carboxamide
Figure imgf000034_0001
0.14 g of 1 -hydroxybenzotriazole, 0.14 cm3 of 2,4-dichlorobenzylamine and 0.14 cm3 of triethylamine are added to 0.2 g of 3-oxo-6-phenyl-2,3-dihydro- pyridazine-4-carboxylic acid prepared as described by Y. Shojiro et al., Chem. Pharm. Bull; 19, (11), p. 2354, in 10 cm3 of dichloromethane. 0.2 g of 1-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride is then added. The mixture is stirred for 48 hours at 19°C. 10 cm3 of distilled water are added. The organic phase is washed again with 3 times 10 cm3 of aqueous normal hydrochloric acid solution and then with 10 cm3 of saturated aqueous sodium chloride solution. The organic phase is then dried over magnesium sulfate. The mixture is filtered through a sinter funnel and then evaporated under reduced pressure (2 kPa; 45°C). The residue is taken up with 10 cm3 of diisopropyl ether. The insoluble material is filtered off through a sinter funnel and then rinsed again with 10 cm3 of diisopropyl ether. After drying (10 kPa; 20°C), 28 mg of N-(2,4- dichlorobenzyl)-3-oxo-6-phenyl-4-yl-2,3-dihydropyridazine-4-carboxamide are obtained in the form of a cream-colored solid melting at about 258°C.
1H NMR spectrum (300 MHz, (CD3) SO d6, δ in ppm): 4.64 (d, J = 6 Hz: 2H); from 7.40 to 7.60 (mt: 5H); 7.66 (broad s: 1 H); 7.92 (mt: 2H); 8.55 (s: 1 H); 10.04 (broad t, J = 6 Hz: 1 H); from 13.80 to 14.15 (broad unresolved peak: 1 H).
[M+1]-peak: 374
Example 2 V-(2,4-dichlorobenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydiropyridazine-4- carboxamide
Figure imgf000035_0001
0.02 cm3 of dimethylformamide and then 0.12 cm3 of oxalyl chloride are added to 0.3 g of 3-oxo-6-(pyridin-4-yl)-2,3-dihydropyridazine-4-carboxylic acid prepared as described in patent FR 2 481 284, dissolved in 10 cm3 of dichloromethane. The reaction medium is stirred for 3 hours at 19°C. A further 0.12 cm3 of oxalyl chloride is then added and the mixture is stirred for a further one hour at 19°C. The reaction mixture is then poured onto a solution of 10 cm3 of dichloromethane containing 0.19 cm3 of triethylamine and 0.21 cm3 of 2,4-dichlorobenzylamine. The reaction medium is stirred for 12 hours at 19°C and then filtered through a sinter funnel, rinsed with 10 cm3 of dichloromethane, 10 cm3 of distilled water and with 10 cm3 of aqueous normal hydrochloric acid solution. After drying (10 kPa; 20°C), 0.25 g of N-(2,4-dichlorobenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4- carboxamide is obtained in the form of a white solid melting at 233°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.64 (d, J = 6 Hz: 2H); 7.45 (mt: 2H); 7.66 (broad s: 1 H); 7.91 (broad d, J = 5 Hz: 2H); 8.62 (s: 1 H); 8.73 (broad d, J = 5 Hz: 2H); 9.95 (broad t, J = 6 Hz: 1 H); 14.25 (unresolved peak: 1 H).
[M+1]-peak: 375,03
Example 3
N-benzyl-3-oxo~6-pyπdin-4-yl-2,3-dihydropyridazine-4-carboxamide
Figure imgf000036_0001
Working as in example 2 for the preparation of N-(2,4-dichlorobenzyl)-3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide, but starting with 0.3 g of 3-oxo- 6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 10 cm3 of dichloromethane, 0.02 cm3 of dimethylformamide, 0.12 cm3 of oxalyl chloride, 0.17 cm3 of benzylamine and 0.19 cm3 of triethylamine, 0.22 g of N-benzyl-3-oxo-6-pyridin-4- yl-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a cream-colored solid melting at 258°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.61 (d, J = 6 Hz: 2H); from 7.25 to 7.45 (mt: 5H); 7.92 (broad d, J = 6 Hz: 2H); 8.64 (s: 1 H); 8.73 (broad d, J = 6 Hz: 2H); 9.93 (broad t, J = 6 Hz: 1 H).
[M+1]-peak: 307 Example 4
N-(4-chlorobenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide
Figure imgf000037_0001
Working as in example 2 for the preparation of N-(2,4-dichlorobenzyl)-3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide, but starting with 0.3 g of 3-oxo- 6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 10 cm3 of dichloromethane, 0.02 cm3 of dimethylformamide, 0.12 cm3 of oxalyl chloride, 0.19 cm3 of 4- chlorobenzylamine and 0.19 cm3 of triethylamine, 0.2 g of N-(4-chlorobenzyl)-3- oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a cream-colored solid melting at 250°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.60 (d, J = 6 Hz: 2H); 7.41 (mt: 4H); 7.92 (broad d, J = 6 Hz: 2H); 8.64 (s: 1 H); 8.73 (broad d, J = 6 Hz: 2H); 9.91 (broad t, J = 6 Hz: 1 H).
[M+1]-peak: 341
Example 5
W-(2-chlorobenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyrϊdazine-4-carboxamide
Figure imgf000037_0002
Working as in example 2 for the preparation of N-(2,4-dichlorobenzyl)-3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide, but starting with 0.3 g of 3-oxo- 6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 10 cm3 of dichloromethane, 0.02 cm3 of dimethylformamide, 0.12 cm3 of oxalyl chloride, 0.19 cm3 of 2- chlorobenzylamine and 0.19 cm3 of triethylamine, 0.25 g of N-(2-chlorobenzyl)-3- oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a white solid melting above 260°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.67 (d, J = 6 Hz: 2H); 7.35 (mt: 2H); from 7.40 to 7.55 (mt: 2H); 7.92 (broad d, J = 6 Hz: 2H); 8.63 (s: 1 H); 8.72 (broad d, J = 6 Hz: 2H); 9.95 (broad t, J = 6 Hz: 1 H); 14.25 (s: 1 H).
[M+1]-peak: 341
Example 6
iV-[2-(2,4-dichlorophenyl)ethyl]-3-oxo-6-pyridin-4-yl-253-dihydropyridazine-4- carboxamide
Figure imgf000038_0001
Working as in example 2 for the preparation of N-(2,4-dichlorobenzyl)-3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide, but starting with 0.3 g of 3-oxo- 6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 10 cm3 of dichloromethane, 0.02 cm3 of dimethylformamide, 0.12 cm3 of oxalyl chloride, 0.23 cm3 of 2,4- dichlorophenylethylamine and 0.19 cm3 of triethylamine, 0.23 g of N-[2-(2,4- dichlorophenyl)ethyl]-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a cream-colored solid melting at 202°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 3.00 (t, J = 7 Hz: 2H); 3.63
(q, J = 7 Hz: 2H); 7.38 (dd, J = 8.5 and 2 Hz: 1 H); 7.44 (d, J = 8 Hz: 1 H); 7.60 (d, J = 2 Hz: 1 H); 7.90 (d mt, J = 6 Hz: 2H); 8.49 (s: 1 H); 8.68 (d mt, J = 6 Hz: 2H); 9.96 (unresolved peak: 1 H); from 13.50 to 14.50 (very broad unresolved peak: 1 H).
[M+1]-peak: 389
Example 7
Λ -(2,4-dichlorophenyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4- carboxamide
Figure imgf000039_0001
Working as in example 2 for the preparation of N-(2,4-dichlorobenzyl)-3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide, but starting with 0.3 g of 3-oxo- 6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 10 cm3 of dichloromethane, 0.02 cm3 of dimethylformamide, 0.12 cm3 of oxalyl chloride, 0.036 g of 2,4- dichloroaniline and 0.19 cm3 of triethylamine, 0.16 g of N-(2,4-dichlorophenyl)-3- oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a cream-colored solid melting above 260°C.
1H NMR spectrum (400 MHz, (CD3)2SO d6 with addition of a few drops of CD3COOD d4, δ in ppm): 7.52 (dd, J = 8.5 and 2.5 Hz: 1 H); 7.75 (d, J = 2.5 Hz: 1 H); 7.96 (d mt, J = 6 Hz: 2H); 8.60 (d, J = 8.5 Hz: 1 H); 8.75 (broad d, J = 6 Hz: 2H); 8.77 (s: 1 H).
[M+1]-peak: 361
Example 8
3-oxo-6-pyridin-4-yl-N-(pyridin-4-ylmethyl)-2,3-dihydropyridazine-4- carboxamide
Figure imgf000040_0001
Working as in example 2 for the preparation of N-(2,4-dichlorobenzyl)-3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide, but starting with 0.3 g of 3-oxo- 6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 10 cm3 of dichloromethane, 0.02 cm3 of dimethylformamide, 0.12 cm3 of oxalyl chloride, 0.15 cm3 of 4- (aminomethyl)pyridine and 0.19 cm3 of triethylamine, 0.14 g of 3-oxo-6-pyridin-4- yl-N-(pyridin-4-ylmethyl)-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a cream-colored solid melting at 254°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.63 (d, J = 6 Hz: 2H); 7.35 (broad d, J = 6 Hz: 2H); 7.92 (d mt, J = 6 Hz: 2H); 8.53 (broad d, J = 6 Hz: 2H); 8.62 (s: 1 H); 8.72 (broad d, J = 6 Hz: 2H); 9.99 (t, J = 6 Hz: 1 H); 14.26 (unresolved peak: 1 H).
[M+1]-peak: 308
Example 9
3-oxo-6-pyridin-4-yl-N-[3-(trifluoromethyl)benzyl]-2,3-dihydropyridazine-4- carboxamide
Figure imgf000040_0002
Working as in example 2 for the preparation of N-(2,4-dichlorobenzyl)-3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide, but starting with 0.3 g of 3-oxo- 6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 10 cm3 of dichloromethane, 0.02 cm3 of dimethylformamide, 0.12 cm3 of oxalyl chloride, 0.21 cm3 of 3- (trifluoromethyl)benzylamine and 0.19 cm3 of triethylamine, 0.22 g of 3-oxo-6- pyridin-4-yl-N-[3-(trifluoromethyl)benzyl]-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a cream-colored solid melting at 224°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.68 (d, J = 6 Hz: 2H); from 7.55 to 7.75 (mt: 3H); 7.74 (broad s: 1 H); 7.91 (d mt, J = 6 Hz: 2H); 8.63 (s: 1 H); 8.72 (d mt, J = 6 Hz: 2H); 9.96 (broad t, J = 6 Hz: 1 H); 14.21 (unresolved peak: 1 H).
[M+1]-peak: 375
Example 10
3-oxo-6-pyridin-4-yl-N-[4-(trifluoromethyl)benzyl]-2,3-dihydropyridazϊne-4- carboxamide
Figure imgf000041_0001
Working as in example 2 for the preparation of N-(2,4-dichlorobenzyl)-3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide, but starting with 0.3 g of 3-oxo- 6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 10 cm3 of dichloromethane, 0.02 cm3 of dimethylformamide, 0.12 cm3 of oxalyl chloride, 0.21 cm3 of 4- (trifluoromethyl)benzylamine and 0.19 cm3 of triethylamine, 0.22 g of 3-oxo-6- pyridin-4-yl-N-[4-(trifluoromethyl)benzyl]-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a cream-colored solid melting at 227°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.69 (d, J = 6 Hz: 2H); 7.59 (broad d, J = 8 Hz: 2H); 7.73 (broad d, J = 8 Hz: 2H); 7.91 (d mt, J = 6 Hz: 2H); 8.62 (s: 1H); 8.72 (d mt, J = 6 Hz: 2H); 10.04 (very broad t, J = 6 Hz: 1 H); 14.24 (unresolved peak: 1H).
[M+1]-peak: 375
Example 11
iV-(3,5-dichlorobenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4- carboxamide
Figure imgf000042_0001
Working as in example 2 for the preparation of N-(2,4-dichlorobenzyl)-3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide, but starting with 0.3 g of 3-oxo- 6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 10 cm3 of dichloromethane, 0.02 cm3 of dimethylformamide, 0.12 cm3 of oxalyl chloride, 0.19 cm3 of 3,5- dichlorobenzylamine and 0.19 cm3 of triethylamine, 0.025 g of N-(3,5- dichlorobenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a white solid melting above 260°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.59 (d, J = 6 Hz: 2H); 7.43 (mt: 2H); 7.51 (mt: 1H); 7.91 (d mt, J = 6 Hz: 2H); 8.57 (s: 1 H); 8.70 (d mt, J = 6 Hz: 2H); 10.14 (unresolved peak: 1 H); 14.18 (broad unresolved peak: 1 H).
[M+1]-peak: 375
Example 12
3-oxo~6-pyridin-4-yl-N-(n-butyl)-2,3-dihydropyridazine-4-carboxamide
Figure imgf000042_0002
Working as in example 2 for the preparation of N-(2,4-dichlorobenzyl)-3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide, but starting with 0.3 g of 3-oxo- 6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 10 cm3 of dichloromethane, 0.02 cm3 of dimethylformamide, 0.12 cm3 of oxalyl chloride, 0.15 cm3 of n- butylamine and 0.19 cm3 of triethylamine, and after purification by chromatography on silica gel (particle size 40-63 μm, under an argon pressure of 150 kPa), eluting with a mixture of dichloromethane and methanol (97.5/2.5 by volume), 0.23 g of 3- oxo-6-pyridin-4-yl-N-(n-butyl)-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a white solid melting at 209°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 0.93 (t, J = 7 Hz: 3H); 1.38 (mt: 2H); 1.55 (mt: 2H); 3.37 (mt: 2H); 7.90 (d mt, J = 6 Hz: 2H); 8.60 (s: 1 H); 8.72 (broad d, J = 6 Hz: 2H); 9.50 (t, J = 6 Hz: 1 H); 14.20 (unresolved peak: 1 H).
[M+1]-peak: 273
Example 13
Ethyl 3-[(3-oxo-6-pyridin-4-yI-2-3-dihydropyridazine-4-carbonyl)amino]propionate
Figure imgf000043_0001
0.733 g of 1 -hydroxybenzotriazole, 0.833 g of β-alanine ethyl ester hydrochloride, 0.96 cm3 of N,N-diisopropylethylamine and 2.06 g of 0-(7-azabenzotriazol-1 -yl)- N,N,N',N'-tetramethyluronium hexafluorophosphate are successively added to 0.94 g of 3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid dissolved in 100 cm3 of N, N-dimethylformamide. The reaction medium is stirred for 12 hours at 19°C. The solvent is evaporated off under reduced pressure (2 kPa; 55°C). The solid residue is triturated in 20 cm3 of dichloromethane, suction-filtered and oven- dried under reduced pressure (10 kPa; 20°C). After purification by chromatography on silica gel (particle size 40-63 μm, under an argon pressure of 150 kPa) eluting with dichloromethane, 0.45 g of ethyl 3-[(3-oxo-6-pyridin-4-yl-2,3- dihydropyridazine-4-carbonyl)amino]propionate is obtained in the form of a white solid melting at 180°C. 1 H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 1.22 (t, J = 7 Hz: 3H); 2.63 (broad t, J = 6.5 Hz: 2H); 3.62 (q, J = 6.5 Hz: 2H); 4.12 (q, J = 7 Hz: 2H); 7.92 (broad d, J = 6 Hz: 2H); 8.61 (s: 1 H); 8.73 (broad d, J = 6 Hz: 2H); 9.69 (broad t, J = 6.5 Hz: 1H).
[M+1]-peak: 317
Example 14
3-oxo-6-pyridin-4-yl-N-(pyridin-3-ylmethyl)-2,3-dihydropyridazine-4-carboxamide
Figure imgf000044_0001
Working as in example 13 for the preparation of ethyl 3-[(3-oxo-6-pyridin-4-yl-2,3- dihydropyridazine-4-carbonyl)amino]propionate, but starting with 0.3 g of 3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 30 cm3 of N,N- dimethylformamide, 0.233 g of 1 -hydroxybenzotriazole, 0.18 cm3 of 3- (aminomethyl)pyridine, 0.31 cm3 of N,N-diisopropylethylamine and 0.65 g of 0-(7- azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate, 0.046 g of 3-oxo-6-pyridin-4-yl-N-(pyridin-3-ylmethyl)-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a yellow solid melting at 262°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.62 (d, J = 6 Hz: 2H); 7.38 (broad dd, J = 8 and 5 Hz: 1 H); 7.79 (very broad d, J = 8 Hz: 1 H); 7.91 (broad d, J = 6 Hz: 2H); 8.48 (broad d, J = 5 Hz: 1 H); 8.60 (broad s: 1 H); 8.62 (s: 1 H); 8.72 (broad d, J = 6 Hz: 2H); 9.94 (broad t, J = 6 Hz: 1 H); 14.15 (unresolved peak: 1 H).
[M+1]-peak: 308
Example 15
3-oxo-6-pyridin-4-yl-N-(pyridin-2-ylmethyl)-2,3-dihydropyridazine-4- carboxamide
Figure imgf000045_0001
Working as in example 13 for the preparation of ethyl 3-[(3-oxo-6-pyridin-4-yl-2,3- dihydropyridazine-4-carbonyl)amino]propionate, but starting with 0.3 g of 3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 30 cm3 of N,N- dimethylformamide, 0.233 g of 1 -hydroxybenzotriazole, 0.18 cm3 of 2- (aminomethyl)pyridine, 0.31 cm3 of N,N-diisopropylethylamine and 0.65 g of 0-(7- azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate, 0.125 g of 3-oxo-6-pyridin-4-yl-N-(pyridin-2-ylmethyl)-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a white solid melting at 242°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.71 (d, J = 6 Hz: 2H); 7.33 (broad dd, J = 8 and 5.5 Hz: 1 H); 7.42 (broad d, J = 8 Hz: 1 H); 7.81 (resolved t, J = 8 and 2 Hz: 1 H); 7.93 (broad d, J = 6 Hz: 2H); 8.57 (broad d, J = 5.5 Hz: 1 H); 8.63 (s: 1H); 8.73 (broad d, J = 6 Hz: 2H); 10.24 (broad t, J = 6 Hz: 1H); from 14.00 to 14.50 (very broad unresolved peak: 1 H).
[M+1]-peak: 308
Example 16
yV-(3,4-dichlorobenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4- carboxamide
Figure imgf000045_0002
Working as in example 13 for the preparation of ethyl 3-[(3-oxo-6-pyridin-4-yl-2,3- dihydropyridazine-4-carbonyl)amino]propionate, but starting with 0.3 g of 3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 30 cm3 of N,N- dimethylformamide, 0.233 g of 1 -hydroxybenzotriazole, 0.19 cm3 of 3,4- dichlorobenzylamine, 0.31 cm3 of N,N-diisopropylethylamine and 0.65 g of 0-(7- azabenzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate, 0.28 g of N-(3,4-dichlorobenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4- carboxamide is obtained in the form of a cream-colored solid melting at 265°C. 1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.58 (d, J = 6 Hz: 2H); 7.37 (dd, J = 8 and 1.5 Hz: 1 H); 7.62 (d, J = 8 Hz: 1 H); 7.64 (mt: 1 H); 7.91 (broad d, J = 6 Hz: 2H); 8.62 (s: 1 H); 8.72 (broad d, J = 6 Hz: 2H); 9.92 (broad t, J = 6 Hz: 1 H); from 14.00 to 14.40 (very broad unresolved peak: 1H).
[M+1]-peak: 375
Example 17
V-(4-morpholin-4-ylbenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4- carboxamide
Figure imgf000046_0001
Working as in example 13 for the preparation of ethyl 3-[(3-oxo-6-pyridin-4-yl-2,3- dihydropyridazine-4-carbonyl)amino]propionate, but starting with 0.3 g of 3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 30 cm3 of N,N- dimethylformamide, 0.233 g of 1 -hydroxybenzotriazole, 0.265 g of 4-morpholino- benzylamine, 0.31 cm3 of N,N-diisopropylethylamine and 0.65 g of 0-(7- azabenzotriazol-1 -yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate, and after purification by chromatography on silica gel (particle size 40-63 μm, under an argon pressure of 150 kPa), eluting with dichloromethane, 0.13 g of N-(4- morpholin-4-ylbenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a yellow solid melting at 252°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 3.09 (t, J = 5 Hz: 4H); 3.74 (t, J = 5 Hz: 4H); 4.48 (d, J = 6 Hz: 2H); 6.93 (d, J = 8 Hz: 2H); 7.24 (d, J = 8 Hz: 2H); 7.91 (broad d, J = 6 Hz: 2H); 8.62 (s: 1H); 8.72 (broad d, J = 6 Hz: 2H); 9.85 (very broad t, J = 6 Hz: 1 H); 14.22 (unresolved peak: 1H). [M+1]-peak: 392,16
Example 18
(Trimethylsilyl)-2-ethoxymethoxy-4-benzonitrile
Figure imgf000047_0001
0.085 g of 4-dimethylaminopyridine and 4.9 cm3 of chloromethylethoxy(trimethylsilyl) are added successively to 3 g of 4- hydroxybenzonitrile dissolved in 60 cm3 of dichloromethane, followed by addition of 5.62 cm3 of triethylamine. The reaction medium is stirred for 12 hours at 19°C and then washed with three times 10 cm3 of aqueous normal hydrochloric acid solution, then with 10 cm3 of water, then with 10 cm3 of normal sodium hydroxide and finally with 10 cm3 of saturated sodium chloride solution. The organic phase is dried over magnesium sulfate, filtered through a sinter funnel and then evaporated under reduced pressure (2 kPa; 45°C). After purification by chromatography on silica gel (particle size 40-63 μm, under an argon pressure of 150 kPa), eluting with dichloromethane, 3.5 g of (trimethylsilyl)-2-ethoxymethoxy-4-benzonitrile are obtained in the form of a colorless oil.
Mass spectrum: El, m/z = 206 (M - SiCH3)+, m/z = 191 (M - Si(CH3)2)+, m/z = 176 (M - Si(CH3)3)+ base peak, m/z = 103 (PhCN)+, m/z = 73 (Si(CH3)3)+
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): - 0.03 (s: 9H); 0.89 (t, J = 8 Hz: 2H); 3.72 (t, J = 8 Hz: 2H); 5.35 (s: 2H); 7.18 (d, J = 9 Hz: 2H); 7.79 (d, J = 9 Hz: 2H).
(Trimethylsilyl)-2-ethoxymethoxy-4-benzylamine
Figure imgf000047_0002
15.5 cm3 of molar lithium aluminum hydride solution are added, at a temperature in the region of 19°C, to 3.5 g of (trimethylsilyl)-2-ethoxymethoxy-4-benzonitrile dissolved in 70 cm3 of tetrahydrofuran. The reaction medium is heated and maintained at the reflux point of the tetrahydrofuran for 4 hours. After cooling to a temperature in the region of 19°C, 0.6 cm3 of water is added to the reaction medium, followed by 0.6 cm3 of aqueous 0.5 N sodium hydroxide solution and 1.8 cm3 of water. The suspension obtained is filtered through a sinter funnel and the residue is washed with 5 times 1.8 cm3 of tetrahydrofuran. The organic phase is dried over magnesium sulfate, filtered through a sinter funnel and then evaporated under reduced pressure (2 kPa; 45°C). 3.3 g of (trimethylsilyl)-2- ethoxymethoxy-4-benzylamine are obtained in the form of a yellow oil.
Mass spectrum: El, m/z = 253 M+, m/z = 194 (M - CH3CH2OCH2)+ base peak, m/z = 180 (M - Si(CH3)3 +, m/z = 73 (Si(CH3)3)+
1H NMR spectrum (400 MHz, (CD3)2SO d6, δ in ppm): 0.00 (s: 9H); 0.90 (t, J = 8 Hz: 2H); 3.66 (s: 2H); 3.71 (t, J = 8 Hz: 2H); 5.20 (s: 2H); 6.95 (broad d, J = 8.5 Hz: 2H); 7.24 (broad d, J = 8.5 Hz: 2H).
N-(4-hydroxybenzyl)-3-oxo-6-pyridin-4-yl-2,3-dϊhydropyridazine-4- carboxamide
Figure imgf000048_0001
Working as in example 13 for the preparation of ethyl 3-[(3-oxo-6-pyridin-4-yl-2,3- dihydropyridazine-4-carbonyl)amino]propionate, but starting with 0.6 g of 3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxylic acid, 60 cm3 of N.N- dimethylformamide, 0.466 g of 1 -hydroxybenzotriazole, 0.91 g of (trimethylsilyl)-2- ethoxymethoxy-4-benzylamine, 0.6 cm3 of N,N-diisopropylethylamine and 1.31 g of 0-(7-azabenzotriazol-1 -yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate, and after purification by high performance liquid chromatography on a 100 x 30 mm HyPURITY® 5 μm column, eluting with a mixture increasing from 25% to 95% of acetonitrile/water (containing 0.05% triftuoroacetic acid), 0.16 g of N-(4- hydroxybenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a cream-colored solid melting at a temperature above 260°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.48 (d, J = 6 Hz: 2H); 6.75 (d, J = 8 Hz: 2H); 7.19 (d, J = 8 Hz: 2H); 8.05 (broad d, J = 6 Hz: 2H); 8.69 (s: 1H); 8.79 (broad d, J = 6 Hz: 2H); from 9.00 to 9.60 (broad unresolved peak: 1 H); 9.74 (t, J = 6 Hz: 1 H); 14.28 (broad s: 1 H).
[M+1]-peak: 323.11
Example 19
4-benzyloxyacetophenone
Figure imgf000049_0001
14.5 cm3 of benzyl bromide and 16.75 g of potassium carbonate are added, at a temperature in the region of 19°C, to 15 g of 4-hydroxyacetophenone dissolved in 180 cm3 of acetone. The reaction medium is heated and maintained at the reflux temperature of the acetone for 4 hours. After cooling to a temperature in the region of 19°C, the reaction medium is suction-filtered through a sinter funnel and the insoluble material is rinsed again with 10 cm3 of acetone. The organic phase is evaporated under reduced pressure (2 kPa; 45°C) and the solid obtained is dissolved in 300 cm3 of ethyl acetate. The organic solution is washed with twice 100 cm3 of water and then with 100 cm3 of saturated sodium chloride solution. The organic phase is dried over magnesium sulfate, filtered through a sinter funnel and then evaporated under reduced pressure (2 kPa; 45°C). The solid residue is triturated in 20 cm3 of pentane, suction-filtered through a sinter funnel and oven- dried under reduced pressure (10 kPa; 20°C). 23.1 g of 4-benzyloxyacetophenone are obtained in the form of a white solid melting at 99°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 2.52 (s: 3H); 5.21 (s: 2H); 7.13 (d, J = 9 Hz: 2H); from 7.30 to 7.55 (mt: 5H); 7.95 (d, J = 9 Hz: 2H). Diethyl hydroxy[2-(4-benzyloxyphenyl)-2-oxoethyl]malonate
Figure imgf000050_0001
19 cm3 of ethyl ketomalonate and 2.5 cm3 of pyridine are added to 23.1 g of 4- benzyloxyacetophenone. The reaction medium is heated and maintained at reflux for 12 hours. After cooling, the reaction medium is purified by chromatography on silica gel (particle size 40-63 μm, under an argon pressure of 150 kPa), eluting with a mixture of dichloromethane and methanol (99/1 by volume). The fractions containing the product are combined and then concentrated under reduced pressure (45°C; 5 kPa). The product obtained is triturated in 150 cm3 of ethanol, filtered through a sinter funnel, washed with twice 50 cm3 of ethanol and 50 cm3 of isopropyl ether to give, after drying under reduced pressure (2 kPa; 55°C), 5.6 g of diethyl hydroxy[2-(4-benzyloxyphenyl)-2-oxoethyl]malonate melting at 80°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 1.21 (t, J = 7 Hz: 6H); 3.65 (s: 2H); 4.18
(q, J = 7 Hz: 4H); 5.22 (s: 2H); 6.25 (s: 1 H); 7.14 (d, J = 9 Hz: 2H); from 7.30 to 7.55 (mt: 5H); 7.94 (d, J = 9 Hz: 2H).
Ethyl 6-[4-(benzyloxy)phenyl]-3-oxo-2,3-dihydropyridazine-4-carboxylate
Figure imgf000050_0002
1.69 g of hydrazine dihydrochloride are added, at a temperature in the region of 19°C, to 5.6 g of diethyl hydroxy[2-(4-benzyloxyphenyl)-2-oxoethyl]malonate dissolved in 180 cm3 of ethanol. The reaction medium is heated and maintained at reflux for 12 hours. After cooling, the solvent is removed under reduced pressure (2 kPa; 55°C). The solid residue is triturated in 20 cm3 of ethanol, suction-filtered through a sinter funnel and oven-dried under reduced pressure (10 kPa; 50°C). 3.85 g of ethyl 6-[4-(benzyloxy)phenyl]-3-oxo-2,3-dihydropyridazine-4-carboxylate are obtained in the form of a green solid melting at 239°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 1.32 (t, J = 7 Hz: 3H); 4.32 (q, J = 7 Hz: 2H); 5.19 (s: 2H); 7.14 (d, J = 9 Hz: 2H); from 7.30 to 7.55 (mt: 5H); 7.84 (d, J = 9 Hz: 2H); 8.30 (s: 1 H); 13.51 (broad s: 1 H).
6-[4-(benzyloxy)phenyl]-3-oxo-2,3,4,5-tetrahydropyridazine-4-carboxylic acid
Figure imgf000051_0001
33 cm3 of one molar sodium hydroxide solution are added to 3.85 g of ethyl 6-[4- (benzy!oxy)phenyl]-3-oxo-2,3-dihydropyridazine-4-carboxylate. The reaction medium is heated and maintained at reflux for 30 minutes. After cooling, 33 cm3 of one molar hydrochloric acid solution are added.
The suspension obtained is filtered through a sinter funnel and the residue is washed with twice 25 cm3 of water and oven-dried under reduced pressure (10 kPa; 50°C). 3.05 g of 6-[4-(benzyloxy)phenyl]-3-oxo-2,3,4,5-tetrahydro- pyridazine-4-carboxylic acid are obtained in the form of a yellow solid melting above 260°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 5.20 (s: 2H); 7.15 (d, J = 9 Hz: 2H); from 7.30 to 7.55 (mt: 5H); 7.92 (d, J = 9 Hz: 2H); 8.42 (s: 1 H). N-(2,4-dichlorobenzyl)-3-oxo-6-[4-(benzyloxy)phenyl]-2,3-dihydropyridazϊne- 4-carboxamide
Figure imgf000052_0001
Working as in example 13 for the preparation of ethyl 3-[(3-oxo-6-pyridin-4-yl-2,3- dihydropyridazine-4-carbonyl)amino]propionate, but starting with 1 g of 6-[4- (benzyloxy)phenyl]-3-oxo-2,3,4,5-tetrahydropyridazine-4-carboxylic acid, 100 cm3 of N, N-dimethylformamide, 0.523 g of 1 -hydroxybenzotriazole, 0.56 cm3 of 2,4- dichlorobenzylamine, 1.1 cm3 of N,N-diisopropylethylamine and 1.47 g of 0-(7- azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate, 1.02 g of N-(2,4-dichlorobenzyl)-3-oxo-6-[4-(benzyloxy)phenyl]-2,3-dihydropyridazine-4- carboxamide are obtained in the form of a yellow solid melting at 225°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.53 (d, J = 6 Hz: 2H); 5.19 (s: 2H); 7.15 (d, J = 9 Hz: 2H); from 7.30 to 7.55 (mt: 7H); 7.66 (broad s: 1 H); 7.86 (d, J = 9 Hz: 2H); 8.47 (s: 1 H); 10.24 (unresolved peak: 1 H); from 13.75 to 13.95 (broad unresolved peak: 1H).
N-benzyl-3-oxo-6-[4-(hydroxy)phenyl]-2,3-dϊhydropyridazine-4-carboxamϊde
Figure imgf000053_0001
0.4 g of N-(2,4-dichlorobenzyl)-3-oxo-6-[4-(benzyloxy)phenyl]-2,3-dihydropyrida- zine-4-carboxamide dissolved in 10 cm3 of methanol. The reaction medium is heated and maintained at reflux for 2 hours. After cooling, the reaction medium is suction-filtered through a sinter funnel and the insoluble material is rinsed again with three times 10 cm3 of hot methanol. The organic phase is evaporated under reduced pressure (2 kPa; 45°C). The residue is recrystallized from methanol. After filtration through a sinter funnel, washing with twice 10 cm3 of methanol and oven- drying under reduced pressure (10 kPa; 50°C), 0.022 g of N-benzyl-3-oxo-6-[4- (hydroxy)phenyl]-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a yellow solid melting at a temperature above 260°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.53 (d, J = 6 Hz: 2H); 6.89 (broad d, J = 9 Hz: 2H); from 7.20 to 7.45 (mt: 5H); 7.75 (broad d, J = 9 Hz: 2H); 8.50 (s: 1 H); 10.02 (t, J = 6 Hz: 1 H).
[M+1]-peak: 322
N-(2,4-dichlorobenzyl)-3-oxo-6-[4-(hydroxy)phenyl]-2-3-dihydropyridazine-4- carboxamide and N-(2,4-dichlorobenzyl)-3-oxo-6-[3-benzyl-4-hydroxyphenyl]- 2,3-dihydropyridazine-4-carboxamide
Figure imgf000054_0001
5 cm3 of trifluoroacetic acid are added to 0.4 g of N-(2,4-dichlorobenzyl)-3-oxo-6- [4-(benzyloxy)phenyl]-2,3-dihydropyridazine-4-carboxamide. The reaction medium is heated and maintained at reflux for 2 hours. After cooling, the solvent is removed under reduced pressure (2 kPa; 55°C). The residue is purified by high performance liquid chromatography on a 100 x 30 mm HyPURITY® 5μm column, eluting with a mixture increasing from 5% to 95% of acetonitrile/water (containing 0.05% trifluoroacetic acid).
0.021 g of N-(2,4-dichlorobenzyl)-3-oxo-6-[4-(hydroxy)phenyl]-2,3-dihydropyri- dazine-4-carboxamide is obtained in the form of a yellow solid melting at a temperature above 260°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.63 (d, J = 6 Hz: 2H); 6.88 (d, J = 8 Hz: 2H); 7.45 (s: 2H); 7.65 (s: 1H); 7.75 (d, J = 8 Hz: 2H); 8.47 (s: 1H); 9.90 (unresolved peak: 1 H); 10.11 (broad t, J = 6 Hz: 1 H); 13.78 (broad unresolved peak: 1H).
0.040 g of N-(2,4-dichlorobenzyl)-3-oxo-6-[3-benzyl-4-hydroxyphenyl]-2,3-dihydro- pyridazine-4-carboxamide is also obtained, in the form of a yellow solid melting at a temperature in the region of 270°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 3.95 (s: 2H); 4.62 (d, J = 6 Hz: 2H); 6.94 (d, J = 8.5 Hz: 1H); from 7.10 to 7.35 (mt: 5H); 7.44 (mt: 2H); 7.60 (dd, J = 8.5 and 2 Hz: 1 H); 7.65 (mt: 2H); 8.45 (s: 1 H); 9.95 (broad s: 1 H); 10.07 (broad t, J = 6 Hz: 1 H); 13.81 (unresolved peak: 1 H). Example 20
Diethyl hydroxy(pyridin-2-oxoethyl)malonate
Figure imgf000055_0001
Working as in example 19 for the preparation of diethyl hydroxy[2-(4- benzyloxyphenyl)-2-oxoethyl]malonate, but starting with 13 cm3 of 2- acetylpyridine, 21 cm3 of ethyl ketomalonate and 2.5 cm3 of pyridine, and after purification by chromatography on silica gel (particle size 40-63 μm, under an argon pressure of 150 kPa), eluting with dichloromethane, 31.1 g of diethyl hydroxy(pyridin-2-oxoethyl)malonate are obtained in the form of a brown oil.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 1.20 (t, J = 7 Hz: 6H); 3.90 (s: 2H); 4.19 (q, J = 7 Hz: 4H); 6.38 (s: 1 H); 7.71 (ddd, J = 7.5 - 5 and 1.5 Hz: 1 H); 7.97 (broad d, J = 7.5 Hz: 1H); 8.04 (resolved t, J = 7.5 and 1.5 Hz: 1H); 8.76 (broad d, J = 5 Hz: 1 H).
Ethyl 6-pyridin-2-yl-3-oxo-2,3-dihydropyridazine-4-carboxylate
Figure imgf000055_0002
Working as in example 19 for the preparation of ethyl 6-[4-(benzyloxy)phenyl]-3- oxo-2,3-dihydropyridazine-4-carboxylate, but starting with 31.1 g of diethyl hydroxy(pyridin-2-oxoethyl)malonate, 11.55 g of hydrazine dihydrochloride and 700 cm3 of ethanol, and after purification by chromatography on silica gel (particle size 40-63 μm, under an argon pressure of 150 kPa), eluting with dichloromethane, followed by recrystallization from ethanol, 6.9 g of ethyl 6- pyridin-2-yl-3-oxo-2,3-dihydropyridazine-4-carboxylate are obtained in the form of a yellow solid melting at 182°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 1.33 (t, J = 7 Hz: 3H); 4.33 (q, J = 7 Hz: 2H); 7.50 (ddd, J = 8 - 5 and 1.5 Hz: 1 H); 7.97 (resolved t, J = 8 and 2 Hz: 1 H); 8.08 (broad d, J = 8 Hz: 1 H); 8.67 (s: 1 H); 8.70 (ddd, J = 5 - 2 and 1.5 Hz: 1H); 13.72 (broad s: 1H).
6-pyridin-2-yl-3-oxo-2.3,4,5-tetrahydropyridazine-4-carboxylic acid
Figure imgf000056_0001
Working as in example 19 for the preparation of 6-[4-(benzyloxy)phenyl]-3-oxo- 2,3,4,5-tetrahydropyridazine-4-carboxylic acid, but starting with 4 g of ethyl 6- pyridin-2-yl-3-oxo-2,3-dihydropyridazine-4-carboxylate, 49 cm3 of one molar sodium hydroxide solution and 50 cm3 of one molar hydrochloric acid solution, 3.44 g of 6-pyridin-2-yl-3-oxo-2,3,4,5-tetrahydropyridazine-4-carboxylic acid are obtained in the form of a beige-colored solid melting at a temperature above 260°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 7.53 (broad dd, J = 7.5 and 5 Hz: 1 H); 7.99 (resolved t, J = 7.5 and 1.5 Hz: 1 H); 8.13 (broad d, J = 7.5 Hz: 1 H); 8.72 (broad d, J = 5 Hz: 1H); 8.83 (s: 1H); from 13.55 to 14.30 (unresolved peak: 2H). iV-(2,4-dichlorobenzyl)-3-oxo-6-pyridin-2-yl-2,3-dihydropyridazine-4- carboxamide
Figure imgf000057_0001
Working as in example 2 for the preparation of N-(2,4-dichlorobenzyl)-3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide, but starting with 0.3 g of 3-oxo- 6-pyridin-2-yl-2,3-dihydropyridazine-4-carboxylic acid, 10 cm3 of dichloromethane, 1 cm3 of dimethylformamide, 0.12 cm3 of oxalyl chloride, 0.21 cm3 of 2,4- dichlorobenzylamine and 0.22 cm3 of triethylamine, 0.22 g of N-(2,4- dichlorobenzyl)-3-oxo-6-pyridin-2-yl-2,3-dihydropyridazine-4-carboxamide is obtained in the form of a cream-colored solid melting at a temperature above 260°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.65 (d, J = 6 Hz: 2H); 7.46 (s: 2H); 7.55 (broad dd, J = 8 and 5 Hz: 1 H); 7.67 (broad s: 1 H); 8.31 (ddd, J = 8 - 2.5 and 2 Hz: 1H); 8.59 (s: 1 H); 8.68 (dd, J = 5 and 2 Hz: 1H); 9.10 (broad d, J = 2.5 Hz: 1 H); 10.09 (t, J = 6 Hz: 1 H).
[M+1]-peak: 375
Example 21
Diethyl hydroxy(pyridin-3-oxoethyl)malonate
Figure imgf000057_0002
Working as in example 19 for the preparation of diethyl hydroxy[2-(4- benzyloxyphenyl)-2-oxoethyl]malonate, but starting with 8 cm3 of 3-acetylpyridine, 14 cm3 of ethyl ketomalonate and 2 cm3 of pyridine, and after purification by chromatography on silica gel (particle size 40-63 μm, under an argon pressure of 150 kPa), eluting with dichloromethane, 8.85 g of diethyl hydroxy(pyridin-3- oxoethyl)malonate are obtained in the form of a brown oil.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 1.21 (t, J = 7 Hz: 6H); 3.76 (s: 2H); 4.20 (q, J = 7 Hz: 4H); 6.44 (s: 1H); 7.59 (broad dd, J = 8 and 5 Hz: 1 H); 8.31 (ddd, J = 8 - 2.5 and 2 Hz: 1H); 8.83 (dd, J = 5 and 2 Hz: 1 H); 9.12 (broad d, J = 2.5 Hz: 1 H).
Ethyl 6-pyridin-3-yl-3-oxo-2,3-dihydropyridazine-4-carboxylate
Figure imgf000058_0001
Working as in example 19 for the preparation of ethyl 6-[4-(benzyloxy)phenyl]-3- oxo-2,3-dihydropyridazine-4-carboxylate, but starting with 8.85 g of diethyl hydroxy(pyridin-3-oxoethyl)malonate, 3.67 g of hydrazine dihydrochloride and 250 cm3 of ethanol, and after recrystallization from ethanol, 3.6 g of ethyl 6-pyridin- 3-yl-3-oxo-2,3-dihydropyridazine-4-carboxylate are obtained in the form of a green solid melting at a temperature in the region of 150°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 1.33 (t, J = 7 Hz: 3H); 4.34 (q, J = 7 Hz: 2H); 7.54 (broad dd, J = 8 and 5 Hz: 1 H); 8.28 (ddd, J = 8 - 2.5 and 2 Hz: 1 H); 8.41 (s: 1 H); 8.67 (dd, J = 5 and 2 Hz: 1 H); 9.09 (broad d, J = 2.5 Hz: 1H); 13.75 (unresolved peak: 1 H).
6-pyridin-3-yl-3-oxo-2,3,4,5-tetrahydropyridazine-4-carboxyIic acid
Figure imgf000059_0001
Working as in example 19 for the preparation of 6-[4-(benzyloxy)phenyl]-3-oxo- 2,3,4,5-tetrahydropyridazine-4-carboxylic acid, but starting with 2 g of ethyl 6- pyridin-3-yl-3-oxo-2,3-dihydropyridazine-4-carboxylate, 24.5 cm3 of one molar sodium hydroxide solution and 25 cm3 of one molar hydrochloric acid solution, 1.65 g of 6-pyridin-3-yl-3-oxo-2,3,4,5-tetrahydropyridazine-4-carboxylic acid are obtained in the form of a cream-colored solid melting at a temperature above 260°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 7.55 (broad dd, J = 8 and 5 Hz: 1H); 8.33 (ddd, J = 8 - 2.5 and 2 Hz; 1H); 8.56 (s: 1H); 8.68 (dd, J = 5 and 2 Hz: 1 H); 9.12 (broad d, J = 2.5 Hz: 1 H); from 13.50 to 14.80 (very broad unresolved peak: 2H).
iV-(2,4-dichlorobenzyl)-3-oxo-6-pyridin-3-yl-2,3-dihydropyridazine-4- carboxamide
Figure imgf000059_0002
Working as in example 2 for the preparation of N-(2,4-dichlorobenzyl)-3-oxo-6- pyridin-4-yl-2,3-dihydropyridazine-4-carboxamide, but starting with 0.3 g of 3-oxo- 6-pyridin-3-yl-2,3-dihydropyridazine-4-carboxylic acid, 10 cm3 of dichloromethane,
1 cm X o -ff d -|;inm.ent.hl .yllffon-rma-iimv.Mide-i, A 0. -1i2 O c Λmr .3 o _f o -,x.,a„l!.y ,l| n cUhllon.rlidne, Λ 0.2-1. c <-.rmv.3 of 2,4- dichlorobenzylamine and 0.22 cm3 of triethylamine, and after purification by chromatography on silica gel (particle size 40-63 μm, under an argon pressure of 150 kPa), eluting with a mixture of dichloromethane and methanol (90/10 by volume), 0.204 g of N-(2,4-dichlorobenzyl)-3-oxo-6-pyridin-3-yl-2,3-dihydro- pyridazine-4-carboxamide is obtained in the form of a cream-colored solid melting at a temperature in the region of 260°C.
1H NMR spectrum (300 MHz, (CD3)2SO d6, δ in ppm): 4.65 (d, J = 6 Hz: 2H); 7.46 (s: 2H); 7.55 (broad dd, J = 8 and 5 Hz: 1H); 7.67 (broad s: 1H); 8.31 (ddd, J = 8 - 2.5 and 2 Hz: 1 H); 8.59 (s: 1 H); 8.68 (dd, J = 5 and 2 Hz: 1 H); 9.10 (broad d, J = 2.5 Hz: 1 H); 10.09 (t, J = 6 Hz: 1 H).
[M+1]-peak: 375
If not stated otherwise, the examples listed in the following table are synthesized according to the above-mentioned methods.
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
©
Ul
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Example 41
6-(4-Hydroxy-3-methoxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid 4-chloro-benzyl amide
a) 3,6-Dichloro-pyridazine-4-carboxylic acid
A solution of 24.9 g 3,6-dichloro-4-methylpyridazine and 56,7 g potassium dichromate in 250 ml of concentrated sulphuric acid are stirred at 40°C for 2 h, the reaction mixture is poured onto 1.5 I ice-water and extracted with ethyl acetate. The organic layer is extracted with water and a saturated solution of NaCl, dried over MgS0 and evaporated. The raw product is used without any further purification.
Yield: 27.1 g
MS:M+1 =193.1
b) 6-Chloro-3-oxo-2-3-dihydro-pyridazine-4-carboxylic acid
27 g of 3,6-dichloro-pyridazine-4-carboxylic acid are stirred at 50°C for 6 h in a 1 : 1 mixture of concentrated sulphuric acid and water. The pure product crystallizes after cooling off the reaction mixture and is filtered.
Yield: 12.48 g
MS:M+1 =175.1
c) 6-Chloro-3-oxo-2,3-dihydro-pyridazine-4-carboxylic acid 4- chloro-benzyl amide
Oxalyl chloride is added to a solution a 8.73 g of 6-chloro-3-oxo-2,3- dihydro-pyridazine-4-carboxylic acid and 1 ml DMF in 250 ml THF at 5- 10°C and the mixture is stirred at room temperature for 2 h. Afterwards, it is evaporated to dryness, the residue dissolved in 450 ml THF and 13.8 g potassium carbonate and a solution of 7.2 g 4-chloro-benzyl amide in THF are added. The solvent is distilled off after 2 h of stirring at room temperature, the residue suspended in 100 ml water and a pH of 6.4 is adjusted. The obtained precipitate is sucked off, suspended again in 50 ml
no water and the pH is adjusted to a. aπerwards, the precipitate is sucked off and dried over phosphorous pentoxide in an exciccator.
Yield: 9.3 g
MS:M+1 =298.
d) 6-Chloro-3-oxo-2-(2-trimethylsilyl-ethoxymethyl)-2,3-dihydro- pyradizine-4-carboxylic acid 4-chloro-benzyl amide
8.6 g of 6-chloro-3-oxo-2,3-dihydro-pyridazine-4-carboxylic acid 4 chloro- benzyl amide are dissolved in 100 ml of absolute DMF and 4.8 g N,N- diisopropylethyl amine are added, then it is stirred at room temperature for 30 min. Afterwards, 5.9 g of trimethylsilylethoxymethyl chloride is added dropwise and it is stirred at room temperature for 5 h. The mixture is added to 1000 ml water for working up, extracted with ethyl acetate, the organic layer is washed with a saturated solution of NaCl and dried over MgS04. After distilling off the solvent the residue is chromatographically purified (silica gel, n-heptane/ethylacetate).
Yield: 7.6 g
MS:M+1 =428.18.
e) 6-(4-Hydroxy-3-methoxy-phenyl)-3-oxo-2-(2-trimethylsilanyl- ethoxymethyl)-2,3-dihydro-pyradizine-4-carboxylic acid 4- chloro-benzyl amide
A solution of 128.5 mg 6-chloro-3-oxo-2-(2-trimethylsilyl-ethoxymethyl)-2,3- dihydropyridazine-4-carboxylic acid 4-chloro-benzyl amide, 91.8 mg 2- methoxy-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-phenol, 82.9 mg K2C03 and 32.1 mg triphenylphosphine in 3.2 ml DME/H20 (2/1) are degassed by conducting of argon. Afterwards, the mixture is diluted with ethyl acetate and washed with 0.5 N of HCI. After drying over MgS04 the solvent is distilled off and the raw product is purified by HPLC (column 125x25, PurospherStar RP18 endcapped, 5 μm; solvent: A: water (0.05 % HCOOH), B: acetonitrile (0.05 % HCOOH)).
Yield: 70.7 mg
in MS:M+1 =516.31
f) 6-(4-Hydroxy-3-methoxy-phenyl)-3-oxo-2,3-dihydro-pyridazine- 4-carboxylic acid 4-chloro-benzyl amide
70.7 mg of 6-(4-hydroxy-3-methoxy-phenyl)-3-oxo-2-(2-trimethylsilanyl- ethoxymethyl)-2,3-dihydro-pyridazine-4-carboxylic acid 4-chloro-benzyl amide and 60 μl ethandithiol and 60 μl Water are dissolved in 750 μl TFA and stirred at room temperature for 3 h. Afterwards, the solvent is distilled off and the raw product purified by HPLC (column 125x25, PurospherStar RP18 endcapped, 5 μm; solvent: A: water (0.05 % HCOOH), B: acetonitrile (0.05 % HCOOH)).
Yield: 19.7 mg
MS:M+1 =386.14
Functional measurements for determination of IC50-values
Tau-phosphorylation
Their activities were determined by measuring the inhibition of phosphorylation of the Tau protein in adult rat cortex sections. The cortex sections having a thickness of 300 μm are prepared from 8-10- week old male OFA rats (Iffa-Credo), sacrificed by decapitation. They are incubated in 5 ml of DMEM medium containing pyruvate and glucose 4.5 g/l at 37°C for 40 min. The sections are then washed twice with the medium, distributed into microtubes (50 μl in 500 μl of medium with or without test compounds), and incubated at 37°C, with stirring. Two hours later, the experiment is stopped by centrifugation. The sections are washed, sonicated and centrifuged at 18300 g, for 15 min at 4°C. The concentration of proteins in supernatant is determined by a commercial assay (BCA Protein Assay, Pierce) based on the Lowry method.
The samples, denatured beforehand for 10 min at 70°C, are separated on a 4-12% Bis-Tris vertical gel in the presence of MOPS-SDS buffer and electrotransferred onto nitrocellulose membrane. Immunolabeling is performed with their monoclonal antibody AD2 which specifically recognizes the phosphorylated epitopes Ser396/404 of the Tau protein. The immunoactive proteins are visualized by adding a second antibody directed against the mouse ιy a s and coupled to peroxidase and a chemiluminescent substrate. The autoradiograms obtained are finally quantified using the 'GeneTools' software from Syngene (GeneGnome, Ozyme) in order to determine an IC50.
The compounds of formula (I) have a very advantageous activity and in particular some compounds have an IC50 of less than 100 μM.
Figure imgf000114_0001
GSK-3β
GSK-β activity is measured using human recombinant GSK-3B and a primed (pre-phosphorylated) substrate peptide (derived from glycogen synthase and containing the phosphorylation sites 3a, b, and c) on basis of the AlphaScreen technology in 384-well plate format (small volume plate, white, GREINER). In a final volume of 11 μl, 2 μl of compound (1 nM-100 mM in kinase buffer, DMSO kept constant at 0.9% ), 2 μl of GSK- 38 solution (0.18 nM) and 2 μl of biot. phospho-glycogen synthase peptide (34 nM) in kinase buffer (20 mM Hepes, pH 7,4, 10 mM MgCI, 200 mM EDTA, 1 mM DTT, 0,1 mg/ml BSA, 10 μM ATP) are incubated at room temperature for 60 min. After adding 2,5 ml Donor beads (20 μg/ml) and 2,5 ml antibody (anti-phospho-glycogen synthase 1 :2000) -coated Acceptor beads (40 μg/ml) in AlphaScreen detection buffer (20 mM Hepes, pH 7,4, 10 mM MgCI, 40 mM EDTA, 1 mM DTT, 0,1 mg/ml BSA , plates are incubated at room temperature (in the dark!) over night and then placed in a reader (Alphaquest or Fusion) to measure final fluorescence. IC50-values are calculated form the fitted curveα corrected for blank values (absence of GSK-3B) and preformed in triplicate.
Figure imgf000115_0001

Claims

1. A compound of formula (I)
Figure imgf000116_0001
wherein A represents A1 or A2
Figure imgf000116_0002
R is unsubstituted or at least monosubstituted CrCio-alkyl, aryl, aryl-(CrCio-alkyl)-, heteroaryl, heteroaryl-(Cι-Cι0-alkyl)-, heterocyclyl, heterocyclyl-(Ci-Cio-alkyl)-, C3-Cι0-cycloalkyl, polycycloalkyl, C2-Cιo-alkenyl or C2-Cι0-alkinyl,
where the substituents are selected from halogen, -CN, CV Cio-alkyl, -N02) -OR1 , -C(0)OR1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -SR1 , -S(0)R1 , -S02R1 , -NHS02R1 , -S02NR1 R2, -C(S)NR1 R2, -NHC(S)R1 , -O-SO2RI , -SO2-O-RI , oxo, -C(0)R1 , -C(NH)NH2, heterocyclyl, C3-Cιo-cycloalkyl, aryl-(CrC6-alkyl)-, aryl, heteroaryl, trifluoromethyl, trifluoromethylsulfanyl and trifluoromethoxy,
and aryl, heterocyclyl and heteroaryl may in turn be at least monosubstituted with Ci-Ce-alkyl, d-Cβ-alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
Ar is unsubstituted or at least monosubstituted aryl or heteroaryl; where the substituents are selected from halogen, -CN, N0 , d-do-alkyl, -0R1 , -C(0)OR1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -NHC(S)R1 , -C(S)NR1 R2, -SR1 , -S(0)R1 , -S02R1 , -NHSO2RI , -S02NR1 R2, -0-S02R1 , -S02- 0-R1 , aryl, heteroaryl, aryl-(Cι-C6-alkyl)-, formyl, trifluoromethyl and trifluoromethoxy,
and aryl and heteroaryl may in turn be at least monosubstituted with Cι-C6-alkyl, CrC6-alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
R1 and R2 are independently from each other
hydrogen;
unsubstituted or at least monosubstituted Ci-Cio-alkyl, C3- Cio-cycloalkyl, aryl, aryl-(Cι-C10-alkyl)-, C2-Cι0-alkenyl, C2- Cio-alkinyl, heterocyclyl, heterocyclyl-(Cι-Cιo-alkyl)- or heteroaryl, where the substituents are selected from halogen, CrC6-alkyl, C C6-alkoxy, CN, N02 , NH2, (Cι-C6-alkyl) amino-, di(CrC6-alkyl)amino-, OH, COOH, -COO-^-Ce- alkyl), -CONH2, formyl, trifluoromethyl and trifluoromethoxy;
heteroaryl is a 5 to 10-membered, aromatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O und S;
aryl is phenyl, indanyl, indenyl or naphthyl;
heterocyclyl is a 5 to 10-membered, aliphatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O and S;
or the racemates, enantiomers, diastereoisomers and mixtures thereof, the tautomers or the physiologically acceptable salts thereof;
with the proviso that A is not -C(0)NH(Cι-C6-alkyl), in case Ar is phenyl which is at least monosubstituted with heterocyclyl or heteroaryl containing nitrogen. A compound according to claim 1 , wherein in formula (I)
A is A1 ;
R is unsubstituted or at least monosubstituted Ci-Cio-alkyl, aryl, aryl-(Ct-Cιo-alkyl)-, heteroaryl, heteroaryl-(Cι-Cιo-alkyl)-, heterocyclyl, heterocyclyl-(Cι-C10-alkyl)-, C3-Cιo-cycloalkyl, polycycloalkyl, C2-Cι0-alkenyl or C2-Cι0-alkinyl,
where the substituents are selected from halogen, -CN, CV do-alkyl, -N02, -OR1 , -C(0)OR1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -SR1 , -S(0)R1 , -S02R1 , -NHS02R1 , -S02NR1 R2, -C(S)NR1 R2, -NHC(S)R1 , -0-S02R1 , -SO2-O-RI , oxo, -C(0)R1 , -C(NH)NH2, heterocyclyl, C3-C10-cycloalkyl, aryl-(CrC6-alkyl)-, aryl, heteroaryl, trifluoromethyl, trifluoromethylsulfanyl and trifluoromethoxy,
and aryl, heterocyclyl and heteroaryl may in turn be at least monosubstituted with Cι-C6-alkyl, d-C6-alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
R1 and R2 independently from each other
hydrogen;
unsubstituted or at least monosubstituted d-Cio-alkyl, C3- Cιo-cycloalkyl, aryl, aryl-(Cι-Cι0-alkyl)-, C2-Cιo-alkenyl, C2- Cio-alkinyl, heterocyclyl, heterocyclyl-(Cι-Cι0-alkyl)- or heteroaryl, where the substituents are selected from halogen, d-C6-alkyl, d-C6-alkoxy, CN, N02 , NH2, (d-C6-alkyl) amino-, di(d-C6-alkyl)amino-, OH, COOH, -COO-(d-C6- alkyl), -CONH2, formyl, trifluoromethyl and trifluoromethoxy;
heteroaryl is a 5 to 10-membered, aromatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O and S; aryl is phenyl, indanyl, indenyl or naphthyl;
heterocyclyl is a 5 to 10-membered, aliphatic, mono- oder bicyclic heterocycle containing one or more heteroatoms selected from N, O and S;
or the racemates, enantiomers, diastereoisomers and mixtures thereof, the tautomers or the physiologically acceptable salts thereof.
3. A compound according to claim 1 or 2, wherein the formula (I)
R is unsubstituted or at least monosubstituted Ci-Cio-alkyl, aryl, aryl-(Cι-Cιo-alkyl)-, heterocyclyl, heterocyclyl-(Ci-Cio-alkyl)-,
C3-Cιo-cycloalkyl, heteroaryl or heteroaryl-(Cι-Cιo-alkyl)-,
where the substituents are selected from halogen, -CN, d- do-Alkyl, -N02, -OR1 , -C(0)OR1 , -0-C(0)R1 , -NR1 R2, -NHC(0)R1 , -C(0)NR1 R2, -SR1 , -S(0)R1 , -S02R1 ,
-NHS02R1 , -S02NR1 R2, -C(S)NR1 R2, -NHC(S)R1 , -0-S02R1 , -SO2-O-RI , oxo, -C(0)R1 , -C(NH)NH2, heterocyclyl, C3-Cι0-cycloalkyl, aryl-(d-C6-alkyl)-, aryl, heteroaryl, trifluoromethyl, trifluoromethylsulfanyl and trifluoromethoxy,
and aryl, heterocyclyl and heteroaryl may in turn be at least monosubstituted with Cι-C6-alkyl, Cι-C6-alkoxy, halogen, trifluoromethyl, trifluoroethoxy or OH;
R1 and R2 are independently from each other
hydrogen;
unsubstituted or at least monosubstituted d-Cio-alkyl, C3-
Cio-cycloalkyl, aryl, aryl-(Crdo-alkyl)-, C2-Cιo-alkenyl, C2- Cio-alkinyl, heterocyclyl, heterocyclyl-(CrCιo-alkyl)- or heteroaryl, where the substituents are selected from halogen, d-Ce-alkyl, C C6-alkoxy, CN, N02 , NH2, (d-C6-alkyl)ami no-, di(d-C6-alkyl)amino-, OH, COOH, -COO- (Ci-Ce-alkyl), -CONH2, formyl, trifluoromethyl and trifluoromethoxy;
heteroaryl is a 5 to 10-membered, aromatic, mono- or bicyclic heterocycle containing one or more heteroatoms selected from N, O and S;
aryl is phenyl, indanyl, indenyl or naphthyl;
heterocyclyl is a 5 to 10-membered, aliphatic, mono- oder bicyclic heterocycle, containing one or more heteroatoms selected from N, O and S;
or the racemates, enantiomers, diastereoisomers and mixtures thereof, the tautomers or the physiologically acceptable salts thereof.
4. A compound according to any of claims 1 to 3, wherein in formula (I)
Ar is unsubstituted or at least monosubstituted phenyl, pyridinyl, pyrimidinyl, pyrazolyl, thiophenyl, isoxazolyl, benzo[b]thiophenyl, benzodioxolyl or thiazolo[3,2-b][1 ,2,4]- tiazolyl,
where the substituents are selected from halogen, -CN, N02, d-Cio-alkyl, -OR1 , -C(0)OR1 , -0-C(0)R1 , -NR1R2, -
NHC(0)R1 , -C(0)NR1 R2, -NHC(S)R1 , -C(S)NR1 R2, -SR1 , -S(0)R1 , -S02R1 , -NHS02R1 , -S02NR1 R2, -0-S02R1 , -S02- 0-R1 , aryl, heteroaryl, aryl-(d-C6-alkyl)-, formyl, trifluoromethyl and trifluoromethoxy,
and aryl and heteroaryl may in turn be at least monosubstituted with Ci-Ce-alkyl, Cι-C6-alkoxy, halogen, trifluoromethyl, trifluoromethoxy or OH;
R1 and R2 are independently from each other
hydrogen; unsubstituted or at least monosubstituted d-Cio-alkyl, C3- Cio-cycloalkyl, aryl, aryl-(Cι-Cι0-alkyl)-, C2-Cι0-alkenyl, C2- Cio-alkinyl, heterocyclyl, heterocyclyl-(Cι-Cι0-alkyl)- or heteroaryl, where the substituents are selected from halogen, Ci-Ce-alkyl, Ci-Ce-alkoxy, CN, N02 , NH2, (Cι-C6-alkyl) amino-, di(d-C6-alkyl)amino-, OH, COOH, -COO-(d-C6- alkyl), -CONH2, formyl, trifluoromethyl and trifluoromethoxy;
heteroaryl is a 5 to 10-membered aromatic, mono- or bicyclic heterocycle, containing one or more heteroatoms selected from N, O and S;
aryl is phenyl, indanyl, indenyl or naphthyl;
heterocyclyl is a 5 to 10-membered aliphatic, mono- oder bicyclic heterocycle, containing one or more heteroatoms selected from N, O and S;
or the racemates, enantiomers, diastereoisomers and mixtures thereof, the tautomers or the physiologically acceptable salts thereof.
5. A compound according to any of claims 1 to 4, wherein in formula (I)
A is A1 ;
R is unsubstituted or at least monosubstituted aryl-(Cι-C6-alkyl)- or heteroaryl-(CrC6-alkyl)-,
where the substituents are selected from halogen, Ci-Cβ- alkyl, -OH, -O-aryl, Ci-Ce-alkoxy, -0-(Ci-C6-alkylen)-N(Ci-C6- alkyl)2, -C(0)OH, -C(0)0-(d-C6-alkyl), -NH2, -N(d-C6-alkyl)2, -NH(Cι-Ce-alkyl), -NH(Cι-Cιo-cycloalkyl), -C(0)NH2, -C(0)NH- heteroaryl, -C(0)NH- (Ci-Ce-alkyl), -S02(d-C6-alkyl), ' -S02NH2, -C(0)-heterocyclyl, -C(NH)NH2, heterocyclyl, aryl-
(Ci-Ce-alkyl)-, aryl, trifluoromethyl, and trifluoromethoxy, and aryl, aeterocyclyl and heteroaryl may in turn be at least monosubstituted with Cι-C3-alkyl, CrC3-alkoxy, fluorine, chlorine, bromine, trifluoromethyl, trifluoromethoxy or OH;
heteroaryl is imidazolyl, thiophenyl, furanyl, isoxazolyl, pyridinyl, pyrimidinyl, benzoimidazolyl, indolyl or benzodioxolyl;
aryl is phenyl or naphthyl;
hetrocyclyl is morpholinyl, piperazinyl or piperidinyl;
or the racemates, enantiomers, diastereoisomers and mixtures thereof, the tautomers or the physiologically acceptable salts thereof.
A compound according to any of claims 1 to 5, wherein in formula (I)
A is A1 ;
Ar is unsubstituted or at least monosubstituted phenyl, pyridin-4- yl or pyrimidin-4-yl,
where the substituents are selected from halogen, Ci-Ce- alkyl, -OH, Cι-C6-alkoxy, -C(0)OH, -C(0)0-(d-C6-alkyl), -NH2, -N(Cι-C6-alkyl)2, -NH (Ci-Ce-alkyl), -NH(d-Cι0- cycloalkyl), -NH(heterocyclyl-(d-C6-alkyl-)), -NH(aryl-(d-C6- alkyl-)), -C(0)NH2, -C(0)NH-(d-C6-alkyl), aryl, and heteroaryl,
and aryl, heterocyclyl and heteroaryl may in turn be at least monosubstituted with d-C3-Alkyl, CrC3-alkoxy, fluorine, chlorine, bromine, trifluoromethyl, trifluoromethoxy or OH;
heteroaryl is pyridinyl or pyrimidinyl;
aryl is phenyl or naphthyl;
hetrocyclyl is morpholinyl, piperazinyl or piperidinyl; or the racemates, enantiomers, diastereoisomers and mixtures thereof, the tautomers or the physiologically acceptable salts thereof.
7. A compound according to any of claims 1 to 6, wherein in formula (I)
A is A1 ;
R is unsubstituted or at least monosubstituted benzyl, phenylethyl-, phenylpropyl-, pyridinylmethyl-, pyridinylethyl- or pyridinylpropyl-,
where the substituents are selected from chlorine, bromine, fluorine, trifluoromethyl and carboxy;
Ar is unsubstituted or at least monosubstituted pyridin-4-yl, pyrimidin-4-yl or phenyl,
where the substituents are selected from methylamino-, ethylamino-, propylamino-, butylamino-, hydroxy, methoxy, ethoxy, methyl, ethyl, propyl, (phenylethyl)amino-, benzylamino- and (morpholinylethyl)amino-;
or the racemates, enantiomers, diastereoisomers and mixtures thereof, the tautomers or the physiologically acceptable salts thereof.
8. A compound according to any of claims 1 to 7 selected from the group consisting of
6-(2-butylamino-pyrimidin-4-yl)-3-oxo-2,3-dihydro-pyridazine-4-car- boxylic acid (3-pyridin- 3-yl-propyl)-amide,
6-(4-hydroxy-3-methoxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid (3-pyridin-3-yl-propyl)-amide,
6-(4-hydroxy-3,5-dimethyl-phenyl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid (3-pyridin-3-yl-propyl)-amide,
6-(4-hydroxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4-carboxylic acid (3- pyridin-3-yl-propyl)-amide, 6-(2-ethylamino-pyrimidin-4-yl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid 4-chloro- benzylamide,
6-(3-chloro-4-hydroxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4-carboxylic acid 4-chloro-benzylamide,
6-(4-hydroxy-3,5-dimethyl-phenyl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid 4-chloro-benzylamide,
4-({[6-(4-hydroxy-3,5-dimethyl-phenyl)-3-oxo-2,3-dihydro-pyridazine-4- carbonyl]-amino}- methyl)-benzoic acid,
4-({[6-(4-hydroxy-3-methoxy-phenyl) -3-0X0-2, 3-dihydro-pyridazine-4- carbonyl]-amino}- methyl)-benzoic acid,
6-(2-butylamino-pyrimidin-4-yl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid (pyridin-3- ylmethyl)-amide,
6-(3-fluoro-4-hydroxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4-carboxylic acid 4-chloro-benzylamide,
6-(4-hydroxy-3-methyl-phenyl)-3-oxo-2,3-dihydro-pyridazine-4-carboxylic acid 4-chloro-benzylamide,
6-[2-(2-morpholin-4-yl-ethylamino)-pyrimidin-4-yl]-3-oxo-2,3-dihydro- pyridazine-4- carboxylic acid 4-chloro-benzylamide,
6-(4-hydroxy-3-methoxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid 4-chloro-benzylamide,
6-(2-methylamino-pyrimidin-4-yl)-3-oxo-2,3-dihydro-pyridazine-4- carboxylic acid 4-chloro-benzylamide,
R-3-oxo-6-[2-(1-phenyl-ethylamino)-pyrimidin-4-yl]-2,3-dihydro- pyridazine-4-carboxylic acid (3-phenyl-propyl)-amide,
6-(4-hydroxy-phenyl)-3-oxo-2,3-dihydro-pyridazine-4-carboxylic acid 4- chloro-benzylamide, 3-oxo-6-pyridin-4-yl-N-[4-(trifluoromethyl)benzyl]-2,3-dihydropyridazine- 4-carboxamide,
3-oxo-6-pyridin-4-yl-2,3-dihydro-pyridazine-4-carboxylic acid 4-bromo- benzylamide,
3-oxo-6-pyridin-4-yl-N-(pyridin-3-ylmethyl)-2,3-dihydropyridazine-4- carboxamide
N-(2,4-dichlorobenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4- carboxamide,
3-oxo-6-pyridin-4-yl-2,3-dihydro-pyridazine-4-carboxylic acid 4-chloro-2- fluoro-benzylamide, and
N-(4-chlorobenzyl)-3-oxo-6-pyridin-4-yl-2,3-dihydropyridazine-4- carboxamide;
or the racemates, enantiomers, diastereoisomers and mixtures thereof, the tautomers or the physiologically acceptable salts thereof.
9. A compound according to any of claims 1 to 8 or a physiologically acceptable salt thereof for use as pharmaceutical.
10. The use of a compound according to any of claims 1 to 8 or a physiologically acceptable salt thereof for the manufacture of a medicament for prophylaxis and/or treatment of diseases in which phosphorylation of the Tau protein is observed.
11. The use of a compound according to any of claims 1 to 8 or a physiologically acceptable salt thereof for the manufacture of a medicament which is an inhibitor of GSK-3β.
12. The use of a compound according to any of claims 1 to 8 or a physiologically acceptable salt thereof for the manufacture of a medicament for prophylaxis and/or treatment of neurodegenerative diseases, strokes, cranial and spinal traumas and peripheral neuropathies, obesity, metabolic diseases, type II diabetes, essential hypertension, atherosclerotic cardiovascular diseases, polycystic ovary syndrome, syndrome X, immunodeficiency or cancer.
13. The use according to claim 12, wherein the neurodegenerative disease is Alzheimer's disease, Parkinson's disease, frontoparietal dementia, corticobasal degeneration or Pick's disease.
14. The use according to claim 12 for prophylaxis and/or treatment of type- 11 -diabetes or Alzheimer's disease.
15. A pharmaceutical preparation comprising an effective dose of at least one compound or a physiologically acceptable salt thereof as defined in any of claims 1 to 8 and a physiologically acceptable carrier.
16. A pharmaceutical preparation according to claim 15, which pharmaceutical preparation is in the form of a pill, tablet, lozenge, coated tablet, granule, capsule, hard or soft gelatin capsule, aqueous solution, alcoholic solution, oily solution, syrup, emulsion suspension, pastille, suppository, solution for injection or infusion, ointment, tincture, cream, lotion, powder, spray, transdermal therapeutic systems, nasal spray, aerosol mixture, microcapsule, implant, rod or plaster.
17. A method for the synthesis of a compound of formula (I) according to any of claims 1 to 8, wherein
a) a compound of formula (IV)
Figure imgf000126_0001
where Y1 is halogen, B(OH)2 or Sn(Cι-Cιo-alkyl) and Y2 is H or a protecting group, is converted with Ar-Z in presence of a palladium complex, where Z is
B(OH)2, B(Cι-C10-alkoxy)2, Sn(d-Cι0-alkyl)3, Zn-(Cι-Cιo-alkyl) or halogen,
or
b) with the proviso that in formula (I) A is A1 , a compound of formula (II)
Figure imgf000127_0001
where X is -OH, CrCio-alkoxy, chlorine or -O-C(O)O-(Cι-Cι0-alkyl), is converted with RNH2 .
PCT/EP2003/012950 2002-11-19 2003-11-19 Pyridazinone derivatives as gsk-3beta inhibitors WO2004046117A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
JP2004552660A JP2006509748A (en) 2002-11-19 2003-11-19 Pyridazinone derivatives as GSK-3 beta inhibitors
CA002506022A CA2506022A1 (en) 2002-11-19 2003-11-19 Pyridazinone derivatives as gsk-3beta inhibitors
DE60315354T DE60315354T2 (en) 2002-11-19 2003-11-19 PYRIDAZINONE DERIVATIVES AS GSK-3BETA HEMMER
AU2003283414A AU2003283414A1 (en) 2002-11-19 2003-11-19 Pyridazinone derivatives as gsk-3beta inhibitors
MXPA05005270A MXPA05005270A (en) 2002-11-19 2003-11-19 Pyridazinone derivatives as gsk-3beta inhibitors.
BR0316720-8A BR0316720A (en) 2002-11-19 2003-11-19 Pyridazinone Derivatives as gsk-3beta Inhibitors
EP03775372A EP1581505B1 (en) 2002-11-19 2003-11-19 Pyridazinone derivatives as gsk-3beta inhibitors
NO20052887A NO20052887L (en) 2002-11-19 2005-06-14 Pyridazinone derivatives such as GSK-3Beta inhibitors.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
FR0214443A FR2847253B1 (en) 2002-11-19 2002-11-19 NOVEL DERIVATIVES OF PYRIDAZINONES AS MEDICAMENTS AND PHARMACEUTICAL COMPOSITIONS COMPRISING THEM
FR0214443 2002-11-19

Publications (1)

Publication Number Publication Date
WO2004046117A1 true WO2004046117A1 (en) 2004-06-03

Family

ID=32187711

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2003/012949 WO2004046130A1 (en) 2002-11-19 2003-11-19 Pyridazinone derivatives as cdk2-inhibitors
PCT/EP2003/012950 WO2004046117A1 (en) 2002-11-19 2003-11-19 Pyridazinone derivatives as gsk-3beta inhibitors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/EP2003/012949 WO2004046130A1 (en) 2002-11-19 2003-11-19 Pyridazinone derivatives as cdk2-inhibitors

Country Status (19)

Country Link
EP (2) EP1581505B1 (en)
JP (1) JP2006509748A (en)
KR (1) KR20050083918A (en)
CN (1) CN1741999A (en)
AR (2) AR042202A1 (en)
AT (2) ATE368651T1 (en)
AU (2) AU2003283414A1 (en)
BR (1) BR0316720A (en)
CA (2) CA2506022A1 (en)
CO (1) CO5700722A2 (en)
DE (2) DE60315516T2 (en)
FR (1) FR2847253B1 (en)
MA (1) MA27814A1 (en)
MX (1) MXPA05005270A (en)
NO (1) NO20052887L (en)
PL (1) PL376129A1 (en)
RU (1) RU2005119151A (en)
TW (2) TW200418478A (en)
WO (2) WO2004046130A1 (en)

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005085230A1 (en) * 2004-03-02 2005-09-15 Sanofi-Aventis Deutschland Gmbh 4-benzimidazol-2-yl-pyridazine-3-one-derivatives, production and use thereof in medicaments
WO2005111018A1 (en) 2004-05-18 2005-11-24 Sanofi-Aventis Deutschland Gmbh Pyridazinone derivatives, methods for producing them and their use as pharmaceuticals
WO2007127475A2 (en) * 2006-04-28 2007-11-08 Northwestern University Pyridazines for demyelinating diseases and neuropathic pain
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
US7470689B2 (en) 2004-03-02 2008-12-30 Aventis Pharma S.A. 4-benzimidazol-2-ylpyridazin-3-one derivatives
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
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
WO2010099217A1 (en) 2009-02-25 2010-09-02 Braincells, Inc. Modulation of neurogenesis using d-cycloserine combinations
WO2010101849A1 (en) * 2009-03-02 2010-09-10 Irm Llc N- (hetero)aryl, 2- (hetero)aryl-substituted acetamides for use as wnt signaling modulators
EP2258357A2 (en) 2005-08-26 2010-12-08 Braincells, Inc. Neurogenesis with acetylcholinesterase inhibitor
EP2275096A2 (en) 2005-08-26 2011-01-19 Braincells, Inc. Neurogenesis via modulation of the muscarinic receptors
US7888357B2 (en) 2001-08-31 2011-02-15 Northwestern University Anti-inflammatory and protein kinase inhibitor compositions and related methods for downregulation of detrimental cellular responses and inhibition of cell death
WO2011023754A1 (en) 2009-08-26 2011-03-03 Sanofi-Aventis Novel crystalline heteroaromatic fluoroglycoside hydrates, pharmaceuticals comprising these compounds and their use
EP2314289A1 (en) 2005-10-31 2011-04-27 Braincells, Inc. Gaba receptor mediated modulation of neurogenesis
WO2011063115A1 (en) 2009-11-19 2011-05-26 Braincells Inc. Combination of nootropic agent with one or more neurogenic or neurogenic sensitizing agents for stimulating or increasing neurogenesis
WO2011091033A1 (en) 2010-01-20 2011-07-28 Braincells, Inc. Modulation of neurogenesis by ppar agents
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
EP2377531A2 (en) 2006-05-09 2011-10-19 Braincells, Inc. Neurogenesis by modulating angiotensin
EP2377530A2 (en) 2005-10-21 2011-10-19 Braincells, Inc. Modulation of neurogenesis by PDE inhibition
US8063047B2 (en) 2004-11-02 2011-11-22 Centre National De La Recherche Scientifique (Cnrs) Pyridazine compounds and methods
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
WO2012003189A1 (en) * 2010-06-29 2012-01-05 Irm Llc Compositions and methods for modulating the wnt signaling pathway
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
US8158627B2 (en) 2006-04-28 2012-04-17 Northwestern University Compositions and treatments using pyridazine compounds and cholinesterase inhibitors
WO2012120058A1 (en) 2011-03-08 2012-09-13 Sanofi Oxathiazine derivatives which are substituted with benzyl or heteromethylene groups, method for producing them, their use as medicine and drug containing said derivatives and the use thereof
WO2012120051A1 (en) 2011-03-08 2012-09-13 Sanofi Benzyl-oxathiazine derivates substituted with adamantane or noradamantane, medicaments containing said compounds 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
WO2012120050A1 (en) 2011-03-08 2012-09-13 Sanofi Novel substituted phenyl-oxathiazine derivatives, method for producing them, drugs containing said compounds 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
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
WO2012120057A1 (en) 2011-03-08 2012-09-13 Sanofi Novel substituted phenyl-oxathiazine derivatives, method for producing them, drugs containing said compounds and the 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
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
US8367672B2 (en) 2004-11-02 2013-02-05 Universite De Strasbourg Pyridazine compounds, compositions and methods
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
AU2012203023C1 (en) * 2009-03-02 2013-11-07 Novartis Ag N-(hetero)aryl, 2- (hetero)aryl-substituted acetamides for use as Wnt signaling modulators
US9408845B2 (en) 2006-04-28 2016-08-09 Northwestern University Formulations containing pyridazine compounds
US10941160B2 (en) 2013-10-18 2021-03-09 Celgene Quanticel Research, Inc. Bromodomain inhibitors
US11040035B2 (en) 2016-05-25 2021-06-22 Bayer Pharma Aktiengesellschaft 3-oxo-2,6-diphenyl-2,3-dihydropyridazine-4-carboxamides

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1598348A1 (en) 2004-05-18 2005-11-23 Aventis Pharma Deutschland GmbH Novel pyridazinone derivatives as inhibitors of CDK2
BRPI0515482A (en) 2004-09-20 2008-07-22 Xenon Pharmaceuticals Inc heterocyclic derivatives and their uses as therapeutic agents
AU2005286793A1 (en) 2004-09-20 2006-03-30 Xenon Pharmaceuticals Inc. Heterocyclic derivatives for the treatment of diseases mediated by stearoyl-CoA desaturase enzymes
CA2580856A1 (en) 2004-09-20 2006-03-30 Xenon Pharmaceuticals Inc. Heterocyclic derivatives and their use as stearoyl-coa desaturase inhibitors
TW200626155A (en) 2004-09-20 2006-08-01 Xenon Pharmaceuticals Inc Heterocyclic derivatives and their use as therapeutic agents
TW200626138A (en) 2004-09-20 2006-08-01 Xenon Pharmaceuticals Inc Heterocyclic derivatives and their use as therapeutic agents
EP1799668A1 (en) 2004-09-20 2007-06-27 Xenon Pharmaceuticals Inc. Heterocyclic derivatives and their use as mediators of stearoyl-coa desaturase
AU2005286647A1 (en) 2004-09-20 2006-03-30 Xenon Pharmaceuticals Inc. Heterocyclic derivatives and their use as stearoyl-CoA desaturase inhibitors
CA2618646A1 (en) 2005-06-03 2007-11-15 Xenon Pharmaceuticals Inc. Aminothiazole derivatives as human stearoyl-coa desaturase inhibitors
US9212146B2 (en) 2008-03-18 2015-12-15 Shanghai Institute Of Materia Medica, Chinese Academy Of Sciences Substituted pyridazinones for the treatment of tumors
CN101538245B (en) 2008-03-18 2011-02-16 中国科学院上海药物研究所 One-class pyridazinone compounds and preparation method and application thereof
GB201201100D0 (en) * 2012-01-20 2012-03-07 Medical Res Council Polypeptides and methods
WO2015127137A1 (en) * 2014-02-19 2015-08-27 Aldea Pharmaceuticals, Inc. Mitochondrial aldehyde dehydrogenase 2 (aldh2) binding polycyclic amides and their use for the treatment of cancer
JOP20190193A1 (en) * 2017-02-09 2019-08-08 Bayer Pharma AG 2-heteroaryl-3-oxo-2,3-dihydropyridazine-4-carboxamides for the treatment of cancer
WO2019101642A1 (en) * 2017-11-21 2019-05-31 Bayer Aktiengesellschaft Sulphur substituted 3-oxo-2,3-dihydropyridazine-4-carboxamides
CN111533697B (en) * 2020-05-07 2023-03-31 成都大学 4-aminopyridazinone compound and preparation method thereof
CN111533735A (en) * 2020-05-08 2020-08-14 张建蒙 Oxo-dihydropyridazine thiazole derivative and application thereof in antitumor drugs
CN111393374A (en) * 2020-05-08 2020-07-10 张建蒙 Oxo-dihydropyridazine derivative and application thereof in antitumor drugs
WO2022113003A1 (en) 2020-11-27 2022-06-02 Rhizen Pharmaceuticals Ag Cdk inhibitors
WO2022149057A1 (en) 2021-01-05 2022-07-14 Rhizen Pharmaceuticals Ag Cdk inhibitors

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5418233A (en) * 1991-10-18 1995-05-23 Karl Thomae Gmbh Heterobiarly derivatives and pharmaceutical compositions thereof
JPH09216883A (en) * 1996-02-09 1997-08-19 Fujisawa Pharmaceut Co Ltd Pyrazolopyridine compound and medicine containing the same compound
EP1061077A1 (en) * 1998-03-02 2000-12-20 Kowa Co., Ltd. Novel pyridazine derivatives and drugs containing the same as the active ingredient
WO2002022587A1 (en) * 2000-09-18 2002-03-21 Eisai Co., Ltd. Pyridazinones and triazinones and medicinal use thereof
WO2002050065A2 (en) * 2000-12-21 2002-06-27 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4127404A1 (en) * 1991-08-19 1993-02-25 Thomae Gmbh Dr K CYCLIC IMINODERIVATES, MEDICAMENTS CONTAINING SUCH COMPOUNDS AND METHOD FOR THE PRODUCTION THEREOF
AU2010399A (en) * 1997-12-24 1999-07-19 Affymetrix, Inc. Methods of using chemical libraries to search for new kinase inhibitors
US6440959B1 (en) * 1999-04-21 2002-08-27 Hoffman-La Roche Inc. Pyrazolobenzodiazepines

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5418233A (en) * 1991-10-18 1995-05-23 Karl Thomae Gmbh Heterobiarly derivatives and pharmaceutical compositions thereof
JPH09216883A (en) * 1996-02-09 1997-08-19 Fujisawa Pharmaceut Co Ltd Pyrazolopyridine compound and medicine containing the same compound
EP1061077A1 (en) * 1998-03-02 2000-12-20 Kowa Co., Ltd. Novel pyridazine derivatives and drugs containing the same as the active ingredient
WO2002022587A1 (en) * 2000-09-18 2002-03-21 Eisai Co., Ltd. Pyridazinones and triazinones and medicinal use thereof
WO2002050065A2 (en) * 2000-12-21 2002-06-27 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
PATENT ABSTRACTS OF JAPAN vol. 1997, no. 12 25 December 1997 (1997-12-25) *

Cited By (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7888357B2 (en) 2001-08-31 2011-02-15 Northwestern University Anti-inflammatory and protein kinase inhibitor compositions and related methods for downregulation of detrimental cellular responses and inhibition of cell death
US8088774B2 (en) 2001-08-31 2012-01-03 Northwestern University Anti-inflammatory and protein kinase inhibitor compositions and related methods for downregulation of detrimental cellular responses and inhibition of cell death
WO2005085230A1 (en) * 2004-03-02 2005-09-15 Sanofi-Aventis Deutschland Gmbh 4-benzimidazol-2-yl-pyridazine-3-one-derivatives, production and use thereof in medicaments
US7754713B2 (en) 2004-03-02 2010-07-13 Aventis Pharma S.A. 4-benzimidazol-2-ylpyridazin-3-one derivatives
US7470689B2 (en) 2004-03-02 2008-12-30 Aventis Pharma S.A. 4-benzimidazol-2-ylpyridazin-3-one derivatives
US7727989B2 (en) 2004-03-02 2010-06-01 Sanofi-Aventis Deutschland Gmbh 4-benzimidazol-2-yl-pyridazine-3-one-derivatives, production and use thereof in medicaments
US7709466B2 (en) 2004-05-18 2010-05-04 Sanofi-Aventis Deutschland Gmbh Pyridazinone derivatives, methods for their production and their use as pharmaceuticals
WO2005111018A1 (en) 2004-05-18 2005-11-24 Sanofi-Aventis Deutschland Gmbh Pyridazinone derivatives, methods for producing them and their use as pharmaceuticals
US8367672B2 (en) 2004-11-02 2013-02-05 Universite De Strasbourg Pyridazine compounds, compositions and methods
US9527819B2 (en) 2004-11-02 2016-12-27 Northwestern University Pyridazine compounds, compositions and methods
US9663493B2 (en) 2004-11-02 2017-05-30 Northwestern University Pyridazine compounds, compositions and methods
US8063047B2 (en) 2004-11-02 2011-11-22 Centre National De La Recherche Scientifique (Cnrs) Pyridazine compounds and methods
US8933076B2 (en) 2004-11-02 2015-01-13 Centre National De La Recherche Scientifique (Cnrs) Pyridazine compounds, compositions and methods
EP2258357A2 (en) 2005-08-26 2010-12-08 Braincells, Inc. Neurogenesis with acetylcholinesterase inhibitor
EP2258359A2 (en) 2005-08-26 2010-12-08 Braincells, Inc. Neurogenesis by muscarinic receptor modulation with sabcomelin
EP2258358A2 (en) 2005-08-26 2010-12-08 Braincells, Inc. Neurogenesis with acetylcholinesterase inhibitor
EP2275096A2 (en) 2005-08-26 2011-01-19 Braincells, Inc. Neurogenesis via modulation of the muscarinic receptors
EP2275095A2 (en) 2005-08-26 2011-01-19 Braincells, Inc. Neurogenesis by muscarinic receptor modulation
EP2377530A2 (en) 2005-10-21 2011-10-19 Braincells, Inc. Modulation of neurogenesis by PDE inhibition
EP2314289A1 (en) 2005-10-31 2011-04-27 Braincells, Inc. Gaba receptor mediated modulation of neurogenesis
US9408845B2 (en) 2006-04-28 2016-08-09 Northwestern University Formulations containing pyridazine compounds
US8158627B2 (en) 2006-04-28 2012-04-17 Northwestern University Compositions and treatments using pyridazine compounds and cholinesterase inhibitors
WO2007127475A3 (en) * 2006-04-28 2008-11-06 Univ Northwestern Pyridazines for demyelinating diseases and neuropathic pain
WO2007127475A2 (en) * 2006-04-28 2007-11-08 Northwestern University Pyridazines for demyelinating diseases and neuropathic pain
EP2377531A2 (en) 2006-05-09 2011-10-19 Braincells, Inc. Neurogenesis by modulating angiotensin
EP2382975A2 (en) 2006-05-09 2011-11-02 Braincells, Inc. Neurogenesis by modulating angiotensin
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
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
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
WO2010099217A1 (en) 2009-02-25 2010-09-02 Braincells, Inc. Modulation of neurogenesis using d-cycloserine combinations
US8546396B2 (en) 2009-03-02 2013-10-01 Irm Llc N-(hetero)aryl, 2-(hetero)aryl—substituted acetamides for use as Wnt signaling modulators
EP2623493A1 (en) * 2009-03-02 2013-08-07 Irm Llc N-(hetero)aryl,2-(hetero)aryl-substituted acetamides for use as WNT signaling modulators
US10251893B2 (en) 2009-03-02 2019-04-09 Novartis Ag N-(hetero)aryl, 2-(hetero)aryl-substituted acetamides for use as Wnt signaling modulators
AU2010221493C1 (en) * 2009-03-02 2013-11-07 Novartis Ag N- (hetero)aryl, 2- (hetero)aryl-substituted acetamides for use as Wnt signaling modulators
AU2012203023C1 (en) * 2009-03-02 2013-11-07 Novartis Ag N-(hetero)aryl, 2- (hetero)aryl-substituted acetamides for use as Wnt signaling modulators
CN102369187A (en) * 2009-03-02 2012-03-07 Irm责任有限公司 N- (hetero)aryl, 2- (hetero)aryl-substituted acetamides for use as wnt signaling modulators
EA021225B1 (en) * 2009-03-02 2015-05-29 Новартис Аг N-(HETERO)ARYL,2-(HETERO)ARYL-SUBSTITUTED ACETAMIDES FOR USE AS Wnt SIGNALING MODULATORS
AU2010221493B2 (en) * 2009-03-02 2012-06-07 Novartis Ag N- (hetero)aryl, 2- (hetero)aryl-substituted acetamides for use as Wnt signaling modulators
JP2012519210A (en) * 2009-03-02 2012-08-23 アイアールエム・リミテッド・ライアビリティ・カンパニー N- (hetero) aryl, 2- (hetero) aryl substituted acetamides for use as WNT signaling regulators
KR101359873B1 (en) 2009-03-02 2014-02-06 아이알엠 엘엘씨 N-(hetero)aryl,2-(hetero)aryl-substituted acetamides for use as wnt signaling modulators
JP2014129381A (en) * 2009-03-02 2014-07-10 Irm Llc N-(hetero)aryl, 2-(hetero)aryl substituted acetamides for using as wnt signaling regulator
US9238646B2 (en) 2009-03-02 2016-01-19 Novartis Ag N-(hetero)aryl, 2-(hetero)aryl-substituted acetamides for use as WNT signaling modulators
WO2010101849A1 (en) * 2009-03-02 2010-09-10 Irm Llc N- (hetero)aryl, 2- (hetero)aryl-substituted acetamides for use as wnt signaling modulators
WO2011023754A1 (en) 2009-08-26 2011-03-03 Sanofi-Aventis Novel crystalline heteroaromatic fluoroglycoside hydrates, pharmaceuticals comprising these compounds and their use
WO2011063115A1 (en) 2009-11-19 2011-05-26 Braincells Inc. Combination of nootropic agent with one or more neurogenic or neurogenic sensitizing agents for stimulating or increasing neurogenesis
WO2011091033A1 (en) 2010-01-20 2011-07-28 Braincells, Inc. Modulation of neurogenesis by ppar agents
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
US9181235B2 (en) 2010-06-29 2015-11-10 Novartis Ag Substituted pyridines for modulating the WNT signaling pathway
AU2011271479B2 (en) * 2010-06-29 2014-08-14 Irm Llc Compositions and methods for modulating the Wnt signaling pathway
WO2012003189A1 (en) * 2010-06-29 2012-01-05 Irm Llc Compositions and methods for modulating the wnt signaling pathway
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
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
WO2012120057A1 (en) 2011-03-08 2012-09-13 Sanofi Novel substituted phenyl-oxathiazine derivatives, method for producing them, drugs containing said compounds and the use thereof
WO2012120051A1 (en) 2011-03-08 2012-09-13 Sanofi Benzyl-oxathiazine derivates substituted with adamantane or noradamantane, medicaments containing said compounds 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
WO2012120050A1 (en) 2011-03-08 2012-09-13 Sanofi Novel substituted phenyl-oxathiazine derivatives, method for producing them, drugs containing said compounds and the 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
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
WO2012120058A1 (en) 2011-03-08 2012-09-13 Sanofi Oxathiazine derivatives which are substituted with benzyl or heteromethylene groups, 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
US10941160B2 (en) 2013-10-18 2021-03-09 Celgene Quanticel Research, Inc. Bromodomain inhibitors
US11884680B2 (en) 2013-10-18 2024-01-30 Celgene Quanticel Research, Inc. Bromodomain inhibitors
US11040035B2 (en) 2016-05-25 2021-06-22 Bayer Pharma Aktiengesellschaft 3-oxo-2,6-diphenyl-2,3-dihydropyridazine-4-carboxamides

Also Published As

Publication number Publication date
WO2004046130A1 (en) 2004-06-03
JP2006509748A (en) 2006-03-23
EP1581505A1 (en) 2005-10-05
EP1611121B1 (en) 2007-08-08
ATE369356T1 (en) 2007-08-15
TW200418478A (en) 2004-10-01
RU2005119151A (en) 2006-01-20
FR2847253A1 (en) 2004-05-21
ATE368651T1 (en) 2007-08-15
EP1611121A1 (en) 2006-01-04
DE60315354D1 (en) 2007-09-13
NO20052887L (en) 2005-07-29
KR20050083918A (en) 2005-08-26
MA27814A1 (en) 2006-04-03
AU2003296586A1 (en) 2004-06-15
CA2506022A1 (en) 2004-06-03
DE60315354T2 (en) 2008-05-08
DE60315516T2 (en) 2008-04-17
AR042089A1 (en) 2005-06-08
CO5700722A2 (en) 2006-11-30
MXPA05005270A (en) 2005-07-25
NO20052887D0 (en) 2005-06-14
BR0316720A (en) 2005-10-18
CN1741999A (en) 2006-03-01
AU2003283414A1 (en) 2004-06-15
EP1581505B1 (en) 2007-08-01
AR042202A1 (en) 2005-06-15
TW200503727A (en) 2005-02-01
FR2847253B1 (en) 2007-05-18
CA2518917A1 (en) 2004-06-03
PL376129A1 (en) 2005-12-12
DE60315516D1 (en) 2007-09-20

Similar Documents

Publication Publication Date Title
WO2004046117A1 (en) Pyridazinone derivatives as gsk-3beta inhibitors
US7727989B2 (en) 4-benzimidazol-2-yl-pyridazine-3-one-derivatives, production and use thereof in medicaments
US7709466B2 (en) Pyridazinone derivatives, methods for their production and their use as pharmaceuticals
JP4792455B2 (en) Novel 4-benzimidazol-2-ylpyridazin-3-one derivatives
KR100814599B1 (en) Imidazole compounds for the treatment of neurodegenerative disorders
US20040142932A1 (en) Substituted pyridazinones
US20050020594A1 (en) Substituted pyridazinones
CZ392597A3 (en) Imidazole compounds, process of their preparation pharmaceutical compositions containing thereof and treatment method
JP6446433B2 (en) Pyrimidine substituted pyrrolidine derivatives, pharmaceutical compositions and uses thereof
US20120289518A1 (en) Derivatives of 2-oxoalkyl-1-piperazin-2-one, preparation method thereof and therapeutic use of same
US7309701B2 (en) Pyridazinone derivatives as pharmaceuticals and pharmaceutical compositions containing them
EP1667679B1 (en) Aminobenzimidazoles as selective melanin concentrating hormone receptor antagonists for the treatment of obesity and related disorders
US7462613B2 (en) Pyridazinone derivatives as pharmaceuticals and pharmaceutical compositions containing them
JP2009529575A (en) Pyridine-containing large heterocyclic compounds as kinase inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref document number: 539916

Country of ref document: NZ

Ref document number: 168517

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 200503825

Country of ref document: ZA

Ref document number: 2506022

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 929/CHENP/2005

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: PA/a/2005/005270

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2004552660

Country of ref document: JP

Ref document number: P20050444A

Country of ref document: HR

Ref document number: 05048069

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 2003283414

Country of ref document: AU

Ref document number: 1020057009083

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: P-2005/0419

Country of ref document: YU

WWE Wipo information: entry into national phase

Ref document number: 20038A50578

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2003775372

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2005119151

Country of ref document: RU

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 376129

Country of ref document: PL

WWP Wipo information: published in national office

Ref document number: 1020057009083

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1-2005-500867

Country of ref document: PH

WWP Wipo information: published in national office

Ref document number: 2003775372

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0316720

Country of ref document: BR

WWG Wipo information: grant in national office

Ref document number: 2003775372

Country of ref document: EP