WO2011121096A1 - PYRAZOL[1,5-a]PYRIMIDINE DERIVATIVES AS WNT PATHWAY ANTAGONISTS - Google Patents

PYRAZOL[1,5-a]PYRIMIDINE DERIVATIVES AS WNT PATHWAY ANTAGONISTS Download PDF

Info

Publication number
WO2011121096A1
WO2011121096A1 PCT/EP2011/055060 EP2011055060W WO2011121096A1 WO 2011121096 A1 WO2011121096 A1 WO 2011121096A1 EP 2011055060 W EP2011055060 W EP 2011055060W WO 2011121096 A1 WO2011121096 A1 WO 2011121096A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
aryl
heteroaryl
halogen
optionally substituted
Prior art date
Application number
PCT/EP2011/055060
Other languages
French (fr)
Inventor
Michael Boutros
Rajendra-Prasad Maskey
Corinna Koch
Florian Fuchs
Sandra Steinbrink
Daniel Gilbert
Original Assignee
Dkfz Deutsches Krebsforschungszentrum
Ruprecht-Karls-Universität Heidelberg
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 Dkfz Deutsches Krebsforschungszentrum, Ruprecht-Karls-Universität Heidelberg filed Critical Dkfz Deutsches Krebsforschungszentrum
Publication of WO2011121096A1 publication Critical patent/WO2011121096A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents

Definitions

  • the present invention relates to compounds having the general formula (I) with the
  • the invention relates to the use of said compounds for modulating of the Wnt signalling pathway activity and their use as a medicament, preferably for the treatment of cancer.
  • the Wnt signalling pathway plays an important role in the regulation of cell proliferation and differentiation. Aberrant activation of the Wnt signalling pathway is known to promote uncontrolled cell growth and survival and can therefore be a major driving force in a broad spectrum of human cancers and diseases. For example, the inhibition of aberrant Wnt signalling pathway activity in cancer cell lines effectively blocks their growth (N. Barker and H. Clevers "Mining the Wnt pathway for cancer therapeutics", Nature Reviews, vol. 5, 2007, pages 997-1014; R. Nusse, "Wnt signalling in disease and in development", Cell Research, Vol. 15, 2005, pages 23-32). Other disorders and diseases are considered to be influenced by an aberrant Wnt signalling pathway, too (see e.g. literature cited above).
  • the Wnt signalling pathway involves a large number of proteins regulating the production of Wnt signalling molecules, their interaction with receptors on target cells and the physiological response of target cells resulting from the exposure of cells to the extra- cellular Wnt ligands.
  • Frizzled (Frz) receptor complexes on the surface of target cells and activate distinct intracellular pathways that are broadly classified as canonical or non-canonical Wnt signalling pathways.
  • the canonical pathway regulates the amount of the protein beta-catenin in a cell and its ability to enter the nucleus of the cell, where it interacts with members of the Tcf/Lef protein family.
  • Beta-catenin and Tcf form active transcription factor complexes in the nucleus and activate the Wnt target gene.
  • the presence of the Tcf-beta-catenin complex in the nucleus is a hallmark in the Wnt signalling pathway indicating its activation.
  • An overview of the Wnt signalling pathway can be found in N. Barker and H. Clevers "Mining the Wnt pathway for cancer therapeutics", Nature Reviews, vol. 5, 2007, pages 997-1014.
  • Tcf-beta-catenin complexes in the nuclei of cells leads to activation of the genetic program considered to promote cancer formation by stimulating cell growth, blocking apoptosis and altering cell movement.
  • the artificial disruption of Tcf-beta-catenin complex formation in colon cancer cells effectively blocks target gene activation and inhibits the growth in vitro.
  • Drugs designed to inhibit the Wnt signalling pathway and consequently the formation of the Tcf-beta-catenin complex in the nucleus of a cell are therefore expected to hold great potential for the treatment of a range of cancers and other diseases associated with the Wnt signalling pathway.
  • An object of the present invention is to provide such compounds. This object is achieved by a compound having the general formula (I)
  • X 1 is CR 1 or N:
  • X 2 is CR 2 or N:
  • X 3 is CR 3 or N:
  • Z is CR 4 or CR 4 R 5 ;
  • Y is N or NR 6 ;
  • L is O or NR 7 ; each A is independently from each other CR 8 R 8 ⁇ which is same or different and one A is optionally selected from the group consisting of O; S; NR 9 ; S(O) and S(0) 2 ;
  • n 2 to 4;
  • R 1 ; R2"; and R 3 J are independently from each other selected from H; OH; halogen;
  • R la ; R lb ; and R lc are independently from each other selected from H; C C 6 alkyl; C 3 - C 7 cycloalkyl; C 3 -C 7 heterocyclyl; C 2 -C 6 alkenyl; C 3 -C 7 aryl; C 3 -C 7 heteroaryl; C8-C 12 biaryl; C8-C 12 heterobiaryl; C 4 -Q5 aralkyl; and C 4 -Q5 heteroarylalkyl; wherein alkyl; cycloalkyl; heterocyclyl; alkenyl; aryl; biaryl; heteroaryl; heterobiaryl; aralkyl; and heteroarylalkyl are optionally substituted with one or more R 10 which are the same or different ; R 10 is selected from halogen, CN, OH, C 1-6 alkyl; C 3 -C 7 cyclyl; C 3 -C 7
  • R 10a N(R 10a )C(O)OR 10b ; and OC(O)N(R 10a R 10b ); wherein cyclyl; heterocyclyl; aryl; heteroaryl; biaryl; heterobiaryl; and alkyl are optionally substituted with one or more R 10a , which are the same or different; R 10a ; R 10b and R 10c are independently from each other selected from H; halogen; C 3 -
  • R 4 is a 5 to 6 membered aromatic or heteroaromatic ring which is optionally substituted by one or more R 4a ; selected from halogen, CN, OH, OR , C(0)R 4B ; C(0)0R 4B ; NR R C(0)NR 4b R 4c ; S(0) 2 N(R 4b R 4c ); S(0)N(R 4b R 4c ); S(0) 2 R 4b ; S(0)R 4b N(R 4b )S(0) 2 N(R 4c R 4d ); SR 4b ; OC(0)R 4b ; N(R 4b )C(0)R 4c ; N(R 4b )S(0) 2 R 4c N(R 4b )S(0)R 4c ; N(R 4b )C(0)N(R 4c R 4d ); N(R 4b )C(0)OR 4c ; OC(0)N(R 4b R 4c ) and CrC 6 alkyl; wherein alkyl is optionally substituted
  • R 1J is selected from halogen, CN, OH, C 1-6 alkyl; OR , C(0)R 1Ja ; C(0)OR 1Ja ;
  • N R i3a R i3b C (0)NR 13a R 13b ; S(0) 2 N(R 13a R 13b ); S(0)N(R 13a R 13b ); S(0) 2 R 13a ; S(0)R 13a ; N(R 13a )S(0) 2 N(R 13b R 13c ); SR 13a ; OC(0)R 13a ; N(R 13a )C(0)R 13b ; N(R 13a )S(0) 2 R 13b ; N(R 13a )S(0)R 13b ; N(R 13a )C(0)N(R 13b R 13c );
  • R 4b , R 4c , R 4d , R 13a ; R 13b and R 13c are independently from each other selected from H; and CrC 6 alkyl; wherein C 1-6 alkyl is optionally substituted with one or more halogen, which are the same or different;
  • R 5 is H or linear CrC 6 alkyl
  • R 6 is selected from H; OH; CrC 6 alkyl; C 3 -C 7 heterocyclyl; C 2 -C 6 alkenyl; C 3 -C 7 aryl; C 3 -C 7 heteroaryl;C 4 -C 1 5 aralkyl; C 4 -C 15 heteroarylalkyl; OR 6a ; C(0)R 6a ; C(0)OR 6a ; OC(0)R 6a ; OC(0)N(R 6a R 6b ); N(R 6a R 6b ); N(R 6a )C(0)N(R 6b R 6c ); and N(R 6a )C(0)OR 6b wherein alkyl; cycloalkyl; heterocyclyl; alkenyl; aryl; heteroaryl; aralkyl; and heteroarylalkyl are optionally substituted by one ore more R 11 , which are the same or different;
  • R 6a ; R 6b ; and R 6c are independently from each other selected from H; C C 6 alkyl; C 3 - C 7 cycloalkyl; C 3 -C 7 heterocyclyl; C 2 -C 6 alkenyl; C 3 -C 7 aryl; C 3 -C 7 heteroaryl; C 4 -C 15 aralkyl; and C 4 -C 15 heteroarylalkyl which are optionally substituted with one or more R 11 , which are the same or different;
  • R 11 is selected from C 1-6 alkyl; halogen, CN; OH; OR lla ; C(0)R lla ; C(0)OR lla N R iia R i ib. C ( 0 )NR lla R llb ; S(0) 2 N(R lla R llb ); S(0)N(R lla R llb ); S(0) 2 R lla S(0)R lla ; N(R lla )S(0) 2 N(R llb R l lc ); SR lla ; OC(0)R llb ; N(R lla )C(0)R llb N(R l la )S(0) 2 R lla ; N(R l la )S(0)R llb ; N(R lla )C(0)N(R llb R llc ):
  • R lla ; R llb ; and R llc are independently from each other selected from H; and C C 6 alkyl; wherein Ci_6 alkyl is optionally substituted with one or more halogen, which are the same or different; R 7 is selected from H; OH; CrC 6 alkyl; C 3 -C 7 cycloalkyl; C 3 -C 7 heterocyclyl; C 2 -
  • R a ; R ; and R c are independently from each other selected from H; C C 6 alkyl; C 3 - C 7 cycloalkyl; C 3 -C 7 heterocyclyl; C 2 -C 6 alkenyl; C 3 -C 7 aryl; C 3 -C 7 heteroaryl; C 4 -Q5 aralkyl; and C 4 -Q5 heteroarylalkyl wherein alkyl; cycloalkyl; heterocyclyl; alkenyl; aryl; heteroaryl; aralkyl; heteroarylalkyl are optionally substituted with one or more R 12 , which are the same or different ;
  • R 1Z is selected from C 1-6 alkyl; halogen, CN, OH, OR 1/a ; C(0)R 1/a ; C(0)OR 1/a NR 12a R 12b ; C(0)NR 12a R 12b ; S(0) 2 N(R 12a R 12b ); S(0)N(R 12a R 12b ); S(0) 2 R 12a
  • N(R 12a )C(0)OR 12b ; and OC(0)N(R 12a R 12b ); wherein C 1-6 alkyl is optionally substituted with one or more halogen, which are the same or different; R a ; R and R c are independently from each other selected from H; and CrC 6 alkyl; wherein C 1-6 alkyl is optionally substituted with one or more halogen; which are the same or different; R 8 ; and R 8 ⁇ i are independently from each other selected from H and C3 ⁇ 4;
  • R 9 is selected from OH; CrC 6 alkyl; C 3 -C 7 cycloalkyl; C 3 -C 7 heterocyclyl; C2-C6 alkenyl; C 3 -C 7 aryl; C 3 -C 7 heteroaryl; C 4 -Q5 aralkyl; C 4 -Q5 heteroarylalkyl; OR 9a ; C(0)R 9a and C(0)OR 9a ; wherein alkyl; cycloalkyl; alkenyl; aryl; aralkyl; and heteroarylalkyl are optionally substituted by one ore more groups selected from OH; halogen; CN; CrC 6 alkyl; OR 9b ;
  • R 9a ; R 9b and R 9c are independently from each other selected from H; and CrC 6 alkyl; wherein C 1-6 alkyl is optionally substituted with one or more halogen; which are the same or different; and/or solvates; hydrates; esters; and pharmaceutically acceptable salts thereof.
  • Alkyl means a linear or branched saturated aliphatic hydrocarbon group e.g. methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, n-pentyl, i-pentyl, n-hexyl, i-hexyl and 3-methyl- pentyl and the like.
  • Alkenyl means a linear or branched unsaturated aliphatic hydrocarbon group with at least one double bond e.g. ethenyl, propenyl (allyl), 1-butenyl, 2-butenyl, 1-pentenyl, 2- pentenyl, 1-hexenyl, 2-hexenyl and 3-hexenyl and the like.
  • Cycloalkyl means a saturated 3 to 8-member hydrocarbon ring e.g. cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • Heterocyclyl means a saturated 3- to 8-member hydrocarbon ring containing 1 to 4 heteroatoms selected from the group N, O and S in the ring. The one or more heteroatoms present in the ring replace a -CH 2 - group in the ring.
  • Preferred C 3 -C 8 heterocyclyl groups containing 1 to 4 heteroatoms selected from the group N, O and S are derived from the following heterocyclic compounds: tetrahydrofurane, pyrrolidine, tetrahydrothiophene, oxazolidine, piperidine, tetrahydropyrane, piperazine, dioxane, morpholine and trioxane.
  • Aryl means an aromatic 3- to 7-numbered hydrocarbon ring e.g. phenyl and the like.
  • Preferred C 3 -C 7 heteroaryl groups containing 1 to 4 heteroatoms selected from the group N, O and S are derived from the following hetero aromatic compounds: Pyrrole, pyrazole, imidazole, triazole, tetrazole, furane, thiophene, oxazole, isoxazole, oxadiazole, thiazole, isothiazole, thiadiazole, pyridine, pyridazine, pyrimidine, pyrazine and triazine.
  • Alkyl means a C 3 -C 7 aryl group substituted with at least one Ci-Cg alkyl group, wherein aryl and alkyl are defined as above.
  • the aralkyl group may be bound via the Ci-Cg alkyl group or via the C 3 -C 7 aryl group.
  • Heteroarylalkyl means a C 3 -C 7 heteroaryl group substituted with at least one Ci-Cg alkyl group, wherein heteroaryl and alkyl are defined as above.
  • the heteroarylalkyl group may be bound via the Ci-Cg alkyl group or via the C 3 -C7 heteroaryl group.
  • Biaryl means an aromatic hydrocarbon with two condensed rings with 8 to 12 carbon atoms.
  • C 8 -C 12 biaryl groups are for example derived from naphthalene and indene.
  • C 8 -C 12 heterobiaryl groups containing 1 to 4 heteroatoms selected from the group N, O and S are for example derived from the following heteroaromatic compounds: indole, benzofurane, benzimidazole, purine, benzothiophen, benzothiazole, isoquinoline, quinoxaline, quinoline and quinazoline,
  • at least one of the group X 1 1 , X2" and X 3 J is N, i.e. at least X 3 is N (Ila), at least X 2 is N (lib) or at least X 1 is N (lie).
  • the compound has one of the following formulae (Ila) to (lie):
  • Y is NR 6
  • Z is CR 4 R 5 and at least one of the group X 1 , X2 and X 3 is N and the compound has the formula (V):
  • Y is NR 6
  • Z is CR 4 R 5 and at least X 3 (Via), at least X 2 (VIb), or at least X 1 (Vic) is N.
  • the compound according to this embodiment has one of the formulae (Via) to (Vic):
  • Y is NR 6
  • Z is CR 4 R 5 and at least X 2 and X 3 are N, at least X 1 and X 3 are N or at least X 1 and X 2 are N.
  • the compound has one of the formulae (Vila) to (VIIc):
  • Y is N
  • Z is CR 4 R 5 and at least X 3 is N (Xal)
  • at least X 2 is N (Xbl) or at least X 1 is N (Xcl) is N or Z is CR 4 and at least X 3 is N (Xa2), at least X 2 is N (Xb2) or at least X 1 is N (Xc2) is N .
  • the compound of this embodiment has one of the formulae (Xal) to (Xc2):
  • Another preferred embodiment of the invention refers to the compound of the formulae (Xlla) to (XIIc) wherein Y is N and at least X 2 and X 3 are N (Xlla), at least X 1 and X 3 are N (Xllb) or at least X 1 and X 2 are N (XIIc):
  • Y is N
  • Z is CR 4 and at least X 2 and X 3 are N (Xllal)
  • at least X 1 and X 3 are N (Xllbl) or at least X 1 and X 2 are N (XIIcl) or
  • Z is CR 4 R 5 and at least X 2 and X 3 are N (XIIa2), at least X 1 and X 3 are N
  • Another preferred embodiment of the invention relates the compound wherein Y, X 1 , and X are N.
  • compound has the formula (XIII):
  • Another preferred embodiment of the invention relates to the compound of formula (XIV) with n is 2:
  • n 3 and the compound has the formula (XV):
  • n 4 and the compound has the formula (XVI):
  • R 4 is selected from C 6 aryl and C 6 heteroaryl and is substituted in p- and both m- positions by three substituents selected independently from each other from R 4a as defined above, wherein the substituents of the two o-positions are preferably H. If Z is CR 4 R 5 it is further preferred that R 5 is H.
  • X 1 is CR 1 with R 1 selected from OH; halogen; Ci-C 6 alkyl; C 3 -C 7 cycloalkyl; C 3 -C 7 heterocyclyl; C 2 -C6 alkenyl; C 3 -C 7 heteroaryl; C4-C 15 aralkyl; C4-C 15
  • R or R is selected from C(0)N(R la )OR lb ; C(0)N(R la )N(R lb )R lc ; and C(0)N(R la )N(R lb )C(0)R lc , and even more preferred R 1 ; R 2 or R 3 is selected from C(0)N(H)OR lb ; C(0)N(H)N(H)R lc ; and C(0)N(H)N(H)C(0)R lc .
  • R 4 is selected from C 6 aryl and C 6 heteroaryl and is substituted in p- and both impositions by three substituents selected independently from each other from R 4a as defined above, wherein the substituents of the two o-positions are preferably H and
  • X 1 is CR 1 with R 1 selected from OH; halogen; Ci-C 6 alkyl; C 3 -C 7 cycloalkyl; C 3 -C 7 heterocyclyl; C2-C6 alkenyl; C 3 -C 7 aryl; C 3 -C 7 heteroaryl; C 4 -C 15 aralkyl; C 4 -C 15 heteroarylalkyl; OR la ; C(0)R la ; C(0)OR la ; C(0)N(R la R lb ); C(0)N(R la )OR lb ; C(0)N(R la )N(R lb )C(0)R lc ; C(0)N(R la )N(R lb )R lc ; S(0) 2 N(R la R lb );
  • R 4 is selected from C 6 aryl and C 6 heteroaryl and is substituted in p- and both i positions by three substituents selected independently from each other from R 4a as defined above, wherein the substituents of the two o-positions are preferably H and
  • R or R is selected from C(0)N(R la )OR lb ; C(0)N(R la )N(R lb )R lc ; and C(0)N(R la )N(R lb )C(0)R lc , and even more preferred R 1 ; R 2 or R 3 is selected from C(0)N(H)OR lb ; C(0)N(H)N(H)R lc ; and C(0)N(H)N(H)C(0)R lc .
  • Described below are exemplary salts of the compounds according to the invention which are included herein.
  • the list of the different salts stated below is not meant to be complete and limiting.
  • Compounds according to the invention which contain one or more acidic groups can be used according to the invention, e.g. as their alkali metal salts, alkaline earth metal salts or ammonium salts. More precise examples of such salts include sodium salts, potassium salts, calcium salts, magnesium salts or salts with ammonia or organic amines such as, e.g. ethylamine, ethanolamine, triethanolamine or amino acids.
  • acids examples include hydrogen chloride, hydrogen bromide, phosphoric acid, sulfuric acid, nitric acid, methanesulfonic acid, p-toluenesulfonic acid, napthalenedisulfonic acid, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propionic acid, pivalic acid, diethylacetic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, malic acid, sulfamic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid and other acids known to a person skilled in the art.
  • the invention also includes, in addition to the salt forms mentioned, inner salts or betaines.
  • the respective salts of the compounds according to the invention can be obtained by customary methods which are known to the person skilled in the art, 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.
  • pharmaceutically acceptable means approved by a regulatory agency such as the EMEA (Europe) and/or the FDA (US) and/or any other national regulatory agency for use in animals, preferably in humans.
  • the invention includes all salts of the compounds according to the invention which, owing to low physiological compatibility, are not directly suitable for use in pharmaceuticals but which can be used, for example, as intermediates for chemical reactions or for the preparation of pharmaceutically acceptable salts or which might be suitable for studying Wnt signalling pathway modulating activity of a compound according of the invention in any suitable manner, such as any suitable in vitro assay.
  • the present invention furthermore includes all solvates of the compounds according to the invention.
  • the present invention furthermore includes derivatives/prodrugs (including the salts thereof) of the compounds according to the invention which contain physiologically tolerable and cleavable groups and which are metabolized in animals, preferably mammals, most preferably humans into a compound according to the invention.
  • the present invention furthermore includes the metabolites of the compounds according to the invention.
  • metabolites refers to all molecules derived from any of the compounds according to the invention in a cell or organism, preferably mammal.
  • metabolites relates to molecules which differ from any molecule which is present in any such cell or organism under physiological conditions.
  • the structure of the metabolites of the compounds according to the invention will be obvious to any person skilled in the art, using the various appropriate methods.
  • the compounds of the invention may be prepared by the three component reaction of an aldehyde R 4 -CHO, wherein R 4 is defined as in formula (I) (e.g. benzaldehyde and substituted benzaldehydes, heteroaromatic aldehydes and substituted heteroaromatic aldehydes), a 5-to-7-membered cyclic 1,3-diketone (e.g.
  • 1,3-cyclohexanone and its derivatives 1,3-cylopentanone and its derivatives, 1,3-cycloheptanone and its derivatives, Thiopyran-3,5-dione and its derivatives, Pyran-3,5-dione and its derivatives, Piperidine- 3,5-dione and its derivatives, Piperidine-2,4-dione and its derivatives, Pyrrolidine-2,4- dione and its derivatives, Furan-2,4-dione and its derivatives), and an amino heteroaromate (e.g.
  • the invention further provides a process for the preparation of the inventive compounds of formulae (I) to (XVI) comprising reacting an aldehyde R 4 -CHO, a 5-to-7-membered cyclic 1,3-diketone and a heteroaromate substituted by NH 2 wherein R 4 is defined as in formula (I).
  • the compounds of the formulae (I) to (XVI) can be purified by customary purification procedures, for example by recrystallization or chromatography.
  • the starting materials for the preparation of the compounds of the formulae (I) to (XVI) are commercially available or can be prepared according to or analogously to literature procedures.
  • the compounds of the invention may serve as a basis for the preparation of the other compounds according to the invention by several methods well known by the person skilled in the art.
  • the present invention relates to the discovery that signal transduction pathways regulated by Wnt can be inhibited, at least in part, by compounds of formulae (I) to (XVI). As set out in more detail below, these compounds can inhibit proliferation of tumor cells having Wnt modulated activity. Therefore, the compounds according to the invention are suited for modulating the Wnt signalling pathway.
  • the term "modulating the Wnt signalling pathway” refers to an effect on the series of events that occur when Wnt proteins bind to cell- surface receptors of the frizzled family resulting in an accumulation of beta-catenin in the cell cytoplasm that reaches the nucleus of a cell, and consequently, the Wnt target genes are expressed.
  • the Wnt signalling pathway may be modulated by direct or indirect modulation.
  • Direct modulation means an interaction of the inventive compounds with proteins directly involved in the Wnt signalling pathway leading to an increase or decrease of the expression of the Wnt target genes.
  • Indirect modulation means an increase or decrease of the expression of the Wnt target genes without a direct interaction of the inventive compound with the components involved in the Wnt signalling pathway. Examples for the indirect modulation of the Wnt signalling pathway are tankyrase-inhibitors and calcium regulators like siperone, thapsigargine and iononycine.
  • Inhibition of tankyrases stabilizes one protein of the Wnt signalling pathway (axin), which, inhibits the Wnt signalling pathway. Inhibition of the decomposition of axin leads to an increase of axine and in turn to the inhibition of the Wnt signalling pathway (Huang et al., Nature 461, pp. 614 to 620 (2009))
  • the activation of a receptor may be the mechanism by which these compounds act as described in US 2007/0219257 Al.
  • the compounds could affect the activity of a Wnt frizzled receptor.
  • the compounds could affect the activity of the serine/ threonine kinase GSK3B, which is involved in the down regulation of B-catenin.
  • the compounds could also affect the activity of the APC gene. In the absence of Wnt signal, the APC protein functions to foster degradation of B-catenin and prevent its nuclear entry.
  • Frizzled Frizzled
  • the pathway can also be activated by mutations of B-catenin that render it resistant to degradation.
  • the compounds could alter the activity of Dishevelled, which is a positive mediator of Wnt signaling.
  • the ability of these compounds to inhibit proliferation of cells may be due to the ability of such molecules to interact with Wnt, or at least to interfere with the ability of those proteins to activate a Wnt-mediated signal transduction pathway.
  • Signal transduction antagonists of different structures, even ones that bind to the same protein in the signaling pathways, may act in slightly different ways. Accordingly, even if a particular condition caused or contributed to by aberrant or unwanted activation of the Wnt pathway shows little response to treatment by one of the antagonists disclosed herein, another of the antagonists disclosed herein may nonetheless be efficacious.
  • One embodiment of the present invention includes the use of compounds of formulae (I) to (XVI) that agonize inhibition of Wnt signaling, such as by inhibiting activation of Wnt downstream components of the signaling pathway, in the regulation of repair and/or functional performance of a wide range of cells, tissues and organs, including normal cells, tissues, and organs.
  • the compounds of formulae (I) to (XVI) have therapeutic and cosmetic applications ranging from regulation of neural tissues, bone and cartilage formation and repair, regulation of spermatogenesis, regulation of smooth muscle, regulation of lung, liver and other organs arising from the primitive gut, regulation of hematopoietic function, regulation of skin and hair growth, etc.
  • the compounds of formula (I) can be applied to cells that are provided in culture (in vitro), or on cells in a whole animal (in vivo).
  • Another embodiment of present invention includes the use of compounds of formulae (I) to (XVI), which antagonize activity of the Wnt pathway resulting in the regulation of repair and/or functional performance of a wide range of cells, tissues, and organs.
  • the inventive compounds have therapeutic and cosmetic applications ranging from regulation of neural tissues, bone and cartilage formation and repair, regulation of spermatogenesis, regulation of smooth muscle, regulation of lung, liver and other organs arising from the primative gut, regulation of hematopoietic function, regulation of skin and hair growth, etc.
  • the compounds of the invention can be applied on cells which are provided in culture (in vitro), or on cells in a whole animal (in vivo).
  • agonist refers to an agent or analog that binds productively to a receptor and mimics its biological activity.
  • antagonist refers to an agent that binds to receptors but does not provoke the normal biological response.
  • an antagonist potentiates or recapitulates, for example, the bioactivity of patched, such as to repress transcription of target genes.
  • Wnt antagonist refers not only to any agent that may act by directly inhibiting the normal function of the Wnt protein, but also to any agent that inhibits the Wnt signaling pathway, and thus recapitulates the function of Wnt.
  • Wnt agonist likewise refers to an agent which antagonizes or blocks the bioactivity of Wnt, such as to increase transcription of target genes.
  • the compounds of the invention are used as Wnt antagonists and used to regulate e.g. proliferation or other biological consequences of mis-expression of Wnt.
  • an elevated Tcf-beta-catenin level in the nucleus of a cell is a hallmark of an aberrant activation of the Wnt signalling pathway and plays a major role in the development of several kinds of cancer.
  • the measurement of the Tcf-beta-catenin level in the nucleus of the cell may be carried out according procedures known to the person skilled in the art.
  • the measurement of the Tcf-beta-catenin level by means of 6xTcF- lucif erase is described below in the experiments.
  • the use of a compound for modulating the Wnt signalling pathway resulting in a decrease of the relative amount of Tcf-beta- catenin complex in the nucleus of a cell is preferred.
  • Modulating the Wnt signalling pathway can be carried out by contacting a cell with a compound according to the invention.
  • said modulation is performed in vitro or in cell culture.
  • in vitro and cell culture assays are available.
  • the modulation can be performed in animals such as mammals.
  • Animal subjects to which the invention is applicable extend to both domestic animals and livestock, raised either as pets or for commercial purposes.
  • Exemplary mammals are mice, rats, guinea pigs, monkeys, dogs and cats.
  • the modulation can also be carried out in humans.
  • the invention also relates to the compounds (I) to (XVI) of the invention for use as a medicament.
  • the compounds are as defined above; furthermore the embodiments as described below with respect to the use as medicament, e.g. formulation, application and combination, also apply to this aspect of the invention.
  • the pharmaceutical preparation or medicaments comprising the compounds of formulae (I) to (XVI) can be effective for both human and animal subjects.
  • Animal subjects to which the invention is applicable extend to both domestic animals and livestock, raised either as pets or for commercial purposes. Examples are dogs, cats, cattle, horses, sheep, hogs, and goats.
  • the invention further relates to the use of the compounds of formulae (I) to (XVI) for modulating the Wnt signalling pathway and to the compounds of formulae (I) to (XVI) for modulating the Wnt signalling pathway.
  • the compounds according to the invention are suited for the use for the preparation of a medicament for modulating the Wnt signalling pathway.
  • the present invention provides pharmaceutical preparations or medicaments comprising a compound such as described herein, formulated in an amount sufficient to regulate, in vivo, Wnt pathway, e.g., proliferation or other biological consequences of mis-expression of Wnt.
  • the invention further relates to the use of a compound according to the invention for the preparation of a medicament for the treatment of a disorder or disease associated with an aberrant activation of Wnt signalling in a mammal and to compounds of the invention for the treatment of a disorder or disease associated with an aberrant activation of Wnt signalling in a mammal.
  • the disorders or diseases associated with the Wnt signalling pathway are for example cell-proliferative disorders, rheumatoid arthritis, diseases connected with aberrant bone density and Dupuytren disease (superficial fibromatosis).
  • a cell proliferation disorder is a disorder which is connected with some degree of abnormal cell proliferation. Especially, cell-proliferation disorders are important for the development of cancer.
  • a further cell-proliferation disorder is proliferative skin disorders which are marked by unwanted or aberrant proliferation of cutaneous tissue, for example X-linked ichtyosis, psoriasis, atopic dermatitis, allergic contact dermatitis, epidermolytic hyperkeratosis and seborrheic dermatitis.
  • the invention relates to the use of the compounds according to the invention for the preparation of a medicament for the treatment of cancer or proliferative skin disorder.
  • the Wnt- signalling pathway is also believed to be involved in the maintenance of stem or progenitor cells in a growing list of adult tissues that includes e.g. skin, blood, gut, prostate, muscle and the nervous system. Stem and progenitor cells are important for cell regeneration and consequently for aging and aging related processes. Therefore, the compounds of the invention are useable for the preparation of a medicament for the treatment of aging and age-related disorders and/or diseases.
  • the compounds of the invention are especially suitable for the use for the preparation of a medicament for the treatment of cancer wherein the cancer is a member of the group multiple myeloma, colon cancer, breast cancer, gastritic cancer, colorectal cancer, lung cancer, prostate cancer, ovarian cancer, bladder cancer, liver cancer, uterine cancer, kidney cancer, leukaemia, gliomas, basal cell carcinoma, rhabdomyosarcoma, mesothelioma, osteosarcoma, medulloblastomas and other primary CNS malignant neuroectodermal tumors.
  • the cancer is a member of the group multiple myeloma, colon cancer, breast cancer, gastritic cancer, colorectal cancer, lung cancer, prostate cancer, ovarian cancer, bladder cancer, liver cancer, uterine cancer, kidney cancer, leukaemia, gliomas, basal cell carcinoma, rhabdomyosarcoma, mesothelioma, osteosarcoma, medullob
  • the compounds of the present invention are applicable to cell culture techniques wherein, whether for genetic or biochemical reasons, the cells have a Wnt receptor.
  • a compound of formulae (I) to (XVI) may be employed in a related method directed towards cells which have a Wnt receptor.
  • In vitro neuronal culture systems have proven to be fundamental and indispensable tools for the study of neural development, as well as the identification of neurotrophic factors such as nerve growth factor (NGF), ciliary trophic factors (CNTF), and brain derived neurotrophic factor (BDNF).
  • NNF nerve growth factor
  • CNTF ciliary trophic factors
  • BDNF brain derived neurotrophic factor
  • One use of the compounds of formulae (I) to (XVI) may be in cultures of neuronal stem cells, such as in the use of such cultures for the generation of new neurons and glia.
  • the cultured cells can be contacted with an aromatic compound of the present invention in order to alter the rate of proliferation of neuronal stem cells in the culture and/or alter the rate of differentiation, or to maintain the integrity of a culture of certain terminally differentiated neuronal cells.
  • the subject method can be used to culture, for example, sensory neurons or, alternatively, motorneurons: Such neuronal cultures can be used as convenient assay systems as well as sources of implantable cells for therapeutic treatments.
  • the compounds of the invention can be used in the treatment of neoplastic or hyperplastic transformations such as may occur in the central nervous system.
  • the compounds can be utilized to cause such transformed cells to become either post-mitotic or apoptotic.
  • the compounds may, therefore, be used as part of a treatment for, e.g., malignant gliomas, meningiomas, medulloblastomas, neuroectodermal tumors, and ependymomas.
  • the compounds of the invention can be used as part of a treatment regimen for malignant meduUoblastoma and other primary CNS malignant neuroectodermal tumors.
  • the compounds of the invention are used as part of treatment program for meduUoblastoma.
  • MeduUoblastoma a primary brain tumor, is the most common brain tumor in children.
  • a meduUoblastoma is a primitive neuroectodermal tumor (PNET) arising in the posterior fossa. Histologically, they are small round cell tumors commonly arranged in true rosettes, but may display some differentiation to astrocytes, ependymal cells or neurons (Rorke; Kleihues). PNET's may arise in other areas of the brain including the pineal gland (pineoblastoma) and cerebrum.
  • PNET neuroectodermal tumor
  • Medulloblastoma/PNET's are known to recur anywhere in the CNS after resection, and can even metastasize to bone. Pretreatment evaluation should therefore include an examination of the spinal cord to exclude the possibility of "dropped metastases".
  • Gadolinium-enhanced MRI has largely replaced myelography for this purpose, and CSF cytology is obtained postoperatively as a routine procedure.
  • the compounds of formulae (I) to (XVI) are used as part of a treatment program for hepatocellular carcinoma.
  • Hepatocellular carcinoma is a form of cancer that arises from hepatocytes, the major cell type of the live, and is one of the most common tumors involving mutations in the Wnt pathway.
  • the compounds of formulae (I) to (XVI) are used as part of treatment program for ependymomas.
  • Ependymomas account for approximately 10% of the pediatric brain tumors in children. Grossly, they are tumors that arise from the ependymal lining of the ventricles and microscopically form rosettes, canals, and perivascular rosettes.
  • Yet another aspect of the present invention concerns the observation in the art that Wnt is involved in morphogenic signals involved in other vertebrate organogenic pathways in addition to neuronal differentiation as described above, having apparent roles in other endodermal patterning, as well as both mesodermal and endodermal differentiation processes.
  • compositions comprising one or more of the inventive compounds can also be utilized for both cell culture and therapeutic uses involving generation and maintenance of non-neuronal tissue.
  • the present invention makes use of the discovery that Wnt is apparently involved in controlling the development of stem cells responsible for formation of the digestive tract, liver, lungs, and other organs which derive from the primitive gut.
  • Shh serves as an inductive signal from the endoderm to the mesoderm, which is critical to gut morphogenesis. Therefore, for example, compounds of formulae (I) to (XVI) can be employed for regulating the development and maintenance of an artificial liver which can have multiple metabolic functions of a normal liver.
  • the compounds of formulae (I) to (XVI) can be used to regulate the proliferation and differentiation of digestive tube stem cells to form hepatocyte cultures which can be used to populate extracellular matrices, or which can be encapsulated in biocompatible polymers, to form both implantable and extracorporeal artificial livers.
  • compositions of inventive compounds can be utilized in conjunction with transplantation of such artificial livers, as well as embryonic liver structures, to regulate uptake of intraperitoneal implantation, vascularization, and in vivo differentiation and maintenance of the engrafted liver tissue.
  • the compounds of formulae (I) to (XVI) can be employed therapeutically to regulate such organs after physical, chemical or pathological insult.
  • therapeutic compositions comprising the compounds of formulae (I) to (XVI) can be utilized in liver repair subsequent to a partial hepatectomy.
  • pancreas and small intestine from the embryonic gut depends on intercellular signalling between the endodermal and mesodermal cells of the gut.
  • the differentiation of intestinal mesoderm into smooth muscle has been suggested to depend on signals from adjacent endodermal cells.
  • Sonic hedgehog One candidate mediator of endodermally derived signals in the embryonic hindgut is Sonic hedgehog. See, for example, Apelqvist et al., Curr. Biol. 7:801-4 (1997).
  • the Shh gene is expressed throughout the embryonic gut endoderm with the exception of the pancreatic bud endoderm, which instead expresses high levels of the homeodomain protein Ipfl/Pdxl (insulin promoter factor 1/pancreatic and duodenal homeobox 1), an essential regulator of early pancreatic development.
  • Ipfl/Pdxl insulin promoter factor 1/pancreatic and duodenal homeobox 1
  • Apelqvist et al., supra have examined whether the differential expression of Shh in the embryonic gut tube controls the differentiation of the surrounding mesoderm into specialised mesoderm derivatives of the small intestine and pancreas. To test this, they used the promoter of the Ipfl/Pdxl gene to selectively express Shh in the developing pancreatic epithelium.
  • pancreatic mesoderm developed into smooth muscle and interstitial cells of Cajal, characteristic of the intestine, rather than into pancreatic mesenchyme and spleen. Also, pancreatic explants exposed to Shh underwent a similar program of intestinal differentiation. These results provide evidence that the differential expression of endodermally derived Shh controls the fate of adjacent mesoderm at different regions of the gut tube.
  • inventive compounds can be used to control or regulate the proliferation and/or differentiation of pancreatic tissue both in vivo and in vitro.
  • the compounds of the present invention may provide therapeutic benefits, with the general strategy being, for example, the correction of aberrant insulin expression, or modulation of differentiation. More generally, however, the present invention relates to the use of the compounds of formulae (I) to (XVI) for inducing and/or maintaining a differentiated state, enhancing survival and/or affecting proliferation of pancreatic cells, by contacting the cells with the subject inhibitors.
  • the compounds of the invention could be used as part of a technique to generate and/or maintain such tissue both in vitro and in vivo.
  • modulation of the function of Wnt can be employed in both cell culture and therapeutic uses involving generation and maintenance ⁇ -cells and possibly also for non- pancreatic tissue, such as in controlling the development and maintenance of tissue from the digestive tract, spleen, lungs, colon, and other organs which derive from the primitive gut.
  • the compounds of the invention can be used in the treatment of hyperplastic and neoplastic disorders effecting pancreatic tissue, particularly those characterized by aberrant proliferation of pancreatic cells.
  • pancreatic cancers are marked by abnormal proliferation of pancreatic cells which can result in alterations of insulin secretory capacity of the pancreas.
  • certain pancreatic hyperplasias such as pancreatic carcinomas, can result in hypoinsulinemia due to dysfunction of ⁇ -cells or decreased islet cell mass.
  • the compounds of the invention can be used to enhance regeneration of the tissue after anti-tumor therapy.
  • the present invention makes use of the apparent involvement of Wnt in regulating the development of pancreatic tissue.
  • the compounds of the invention can be employed therapeutically to regulate the pancreas after physical, chemical or pathological insult.
  • the subject method can be applied to cell culture techniques, and in particular, may be employed to enhance the initial generation of prosthetic pancreatic tissue devices. Manipulation of proliferation and differentiation of pancreatic tissue, for example, by altering Wnt, can provide a means for more carefully controlling the characteristics of a cultured tissue.
  • manipulation of the differentiative state of pancreatic tissue can be utilized in conjunction with transplantation of artificial pancreas so as to promote implantation, vascularization, and in vivo differentiation and maintenance of the engrafted tissue.
  • manipulation of Wnt function to affect tissue differentiation can be utilized as a means of maintaining graft viability.
  • the compounds of the invention can be used to regulate regeneration of lung tissue, e.g., in the treatment of emphysema.
  • tumors may, based on evidence such as involvement of the Wnt pathway in these tumors, or detected expression of Wnt or its receptors in these tissues during development, be affected by treatment with the compounds of the invention.
  • Such tumors include, but are by no means limited to, tumors related to Gorlin's syndrome (e.g., basal cell carcinoma, meduUoblastoma, meningioma, etc.), tumors evidenced in pet knock-out mice (e.g., hemangioma, rhabdomyosarcoma, etc.), tumors resulting from gli-1 amplification (e.g., glioblastoma, sarcoma, etc.), tumors connected with TRC8, a ptc homolog (e.g., renal carcinoma, thyroid carcinoma, etc.), Ext-l-related tumors (e.g., bone cancer, etc.), Shh-induced tumors (e.g., lung cancer, chon-drosarcomas, etc.),
  • compositions comprising one or more of the compounds of the invention can be used in the in vitro generation of skeletal tissue, such as from skeleto genie stem cells, as well as the in vivo treatment of skeletal tissue deficiencies.
  • the present invention particularly contemplates the use of compounds of the invention to regulate the rate of chondrogenesis and/or osteogenesis.
  • skeletal tissue deficiency it is meant a deficiency in bone or other skeletal connective tissue at any site where it is desired to restore the bone or connective tissue, no matter how the deficiency originated, e.g. whether as a result of surgical intervention, removal of tumor, ulceration, implant, fracture, or other traumatic or degenerative conditions.
  • the compounds of the invention can be used as part of a regimen for restoring cartilage function to a connective tissue. They are useful in, for example, the repair of defects or lesions in cartilage tissue which is the result of degenerative wear such as that which results in arthritis, as well as other mechanical derangements which may be caused by trauma to the tissue, such as a displacement of torn meniscus tissue, meniscectomy, a laxation of a joint by a torn ligament, malignment of joints, bone fracture, or by hereditary disease.
  • the compounds of the invention may also be useful for remodeling cartilage matrix, such as in plastic or reconstructive surgery, as well as periodontal surgery. They may also be applied to improving a previous reparative procedure, for example, following surgical repair of a meniscus, ligament, or cartilage. Furthermore, it may prevent the onset or exacerbation of degenerative disease if applied early enough after trauma.
  • One embodiment of the present invention relates to the treating of the afflicted connective tissue with a therapeutically effective amount of compounds of the invention to regulate a cartilage repair response in the connective tissue by managing the rate of differentiation and/or proliferation of chondrocytes embedded in the tissue.
  • connective tissues as articular cartilage, interarticular cartilage (menisci), costal cartilage (connecting the true ribs and the sternum), ligaments, and tendons are particularly amenable to treatment in reconstructive and/or regenerative therapies using the subject method.
  • regenerative therapies include treatment of degenerative states which have progressed to the point of which impairment of the tissue is obviously manifest, as well as preventive treatments of tissue where degeneration is in its earliest stages or imminent.
  • the compounds of the invention can be used as part of a therapeutic intervention in the treatment of cartilage of a diarthroidal joint, such as a knee, an ankle, an elbow, a hip, a wrist, a knuckle of either a finger or toe, or a tempomandibular joint.
  • the treatment can be directed to the meniscus of the joint, to the articular cartilage of the joint, or both.
  • the compounds of the invention can be used to treat a degenerative disorder of a knee, such as which might be the result of traumatic injury (e.g., a sports injury or excessive wear) or osteoarthritis.
  • the compounds of the invention may be administered as an injection into the joint with, for instance, an arthroscopic needle.
  • the injected agent can be in the form of a hydrogel or other slow release vehicle described above in order to permit a more extended and regular contact of the agent with the treated tissue.
  • the present invention further contemplates the use of the compounds of the invention in the field of cartilage transplantation and prosthetic device therapies.
  • problems arise, for instance, because the characteristics of cartilage and fibrocartilage vary between different tissue: such as between articular, meniscal cartilage, ligaments, and tendons, between the two ends of the same ligament or tendon, and between the superficial and deep parts of the tissue.
  • the zonal arrangement of these tissues may reflect a gradual change in mechanical properties, and failure occurs when implanted tissue, which has not differentiated under those conditions, lacks the ability to appropriately respond. For instance, when meniscal cartilage is used to repair anterior cruciate ligaments, the tissue undergoes a metaplasia to pure fibrous tissue.
  • the compounds of the invention can be used to particularly address this problem, by helping to adaptively control the implanted cells in the new environment and effectively resemble hypertrophic chondrocytes of an earlier developmental stage of the tissue.
  • the compounds of the invention can be applied to enhancing both the generation of prosthetic cartilage devices and to their implantation.
  • the need for improved treatment has motivated research aimed at creating new cartilage that is based on collagen- glycosaminogly-can templates (Stone et al., Clin. Orthop. Relat. Red 252: 129 (1990)), isolated chondrocytes (Grande et al., J. Orthop. Res.
  • chondrocytes can be grown in culture on biodegradable, biocompatible highly porous scaffolds formed from polymers such as polyglycolic acid, polylactic acid, agarose gel, or other polymers which degrade over time as function of hydrolysis of the polymer backbone into innocuous monomers.
  • the matrices are designed to allow adequate nutrient and gas exchange to the cells until engraftment occurs.
  • the cells can be cultured in vitro until adequate cell volume and density has developed for the cells to be implanted.
  • the matrices can be cast or molded into a desired shape on an individual basis, so that the final product closely resembles the patient's own ear or nose (by way of example), or flexible matrices can be used which allow for manipulation at the time of implantation, as in a joint.
  • the implants are contacted with a subject aromatic compound during certain stages of the culturing process in order to manage the rate of differentiation of chondrocytes and the formation of hypertrophic chrondrocytes in the culture.
  • the implanted device is treated with a compound of formulae (I) to (XVI) in order to actively remodel the implanted matrix and to make it more suitable for its intended function.
  • the artificial transplants suffer from the same deficiency of not being derived in a setting which is comparable to the actual mechanical environment in which the matrix is implanted.
  • the ability to regulate the chondrocytes in the matrix by the subject method can allow the implant to acquire characteristics similar to the tissue for which it is intended to replace.
  • the compounds of the invention are used to enhance attachment of prosthetic devices.
  • the compounds of the invention can be used in the implantation of a periodontal prosthesis, wherein the treatment of the surrounding connective tissue stimulates formation of periodontal ligament about the prosthesis.
  • the compounds of the invention can be employed as part of a regimen for the generation of bone (osteogenesis) at a site in the animal where such skeletal tissue is deficient.
  • Indian hedgehog (Ihh) is particularly associated with the hypertrophic chondrocytes that are ultimately replaced by osteoblasts.
  • administration of a compound of the present invention can be employed as part of a method for regulating the rate of bone loss in a subject.
  • preparations comprising subject compounds can be employed, for example, to control endoch-ondral ossification in the formation of a "model" for ossification.
  • the compounds of the invention also have wide applicability to the treatment or prophylaxis of disorders afflicting epithelial tissue, as well as in cosmetic uses. In general, this includes a step of administering to an animal an amount of a subject aromatic compound effective to alter the growth state of a treated epithelial tissue.
  • the mode of administration and dosage regimens will vary depending on the epithelial tissue(s) which is to be treated. For example, topical formulations will be preferred where the treated tissue is epidermal tissue, such as dermal or mucosal tissues.
  • scarring can be an important obstacle in regaining normal function and appearance of healed skin. This is particularly true when pathologic scarring such as keloids or hypertrophic scars of the hands or face causes functional disability or physical deformity. In the severest circumstances, such scarring may precipitate psychosocial distress and a life of economic deprivation.
  • Wound repair includes the stages of hemostasis, inflammation, proliferation, and remodeling. The proliferative stage involves multiplication of fibroblasts and endothelial and epithelial cells. Through the use of the compounds of the invention, the rate of proliferation of epithelial cells in and proximal to the wound can be controlled in order to accelerate closure of the wound and/or minimize the formation of scar tissue.
  • the present treatment can also be effective as part of a therapeutic regimen for treating oral and paraoral ulcers, e.g., resulting from radiation and/or chemotherapy.
  • Such ulcers commonly develop within days after chemotherapy or radiation therapy.
  • These ulcers usually begin as small, painful irregularly shaped lesions usually covered by a delicate gray necrotic membrane and surrounded by inflammatory tissue.
  • a lack of treatment results in proliferation of tissue around the periphery of the lesion on an inflammatory basis.
  • the epithelium bordering the ulcer usually demonstrates proliferative activity, resulting in loss of continuity of surface epithelium.
  • a treatment for such ulcers which includes application of a compound of formulae (I) to (XVI) can reduce the abnormal proliferation and differentiation of the affected epithelium, helping to reduce the severity of subsequent inflammatory events.
  • the compounds of the invention and compositions thereof can also be used to treat wounds resulting from dermatological diseases, such as lesions resulting from autoimmune disorders such as psoriasis.
  • Atopic dermititis refers to skin trauma resulting from allergies associated with an immune response caused by allergens such as pollens, foods, dander, insect venoms and plant toxins.
  • antiproliferative preparations of subject compounds can be used to inhibit lens epithelial cell proliferation to prevent post-operative complications of extracapsular cataract extraction.
  • Cataract is an intractable eye disease and various studies on a treatment of cataract have been made. But at present, the treatment of cataract is attained by surgical operations. Cataract surgery has been applied for a long time and various operative methods have been examined. Extracapsular lens extraction has become the method of choice for removing cataracts. The major medical advantages of this technique over intra-capsular extraction are lower incidence of aphakic cystoid macular edema and retinal detachment. Extracapsular extraction is also required for implantation of posterior chamber type intraocular lenses which are now considered to be the lenses of choice in most cases.
  • a disadvantage of extracapsular cataract extraction is the high incidence of posterior lens capsule opacification, often called after-cataract, which can occur in up to 50% of cases within three years after surgery.
  • After-cataract is caused by proliferation of equatorial and anterior capsule lens epithelial cells which remain after extracapsular lens extraction. These cells proliferate to cause Sommerling rings, and along with fibroblasts which also deposit and occur on the posterior capsule, cause opacification of the posterior capsule, which interferes with vision. Prevention of after-cataract would be preferable to treatment.
  • the present invention provides a means for inhibiting proliferation of the remaining lens epithelial cells.
  • such cells can be induced to remain quiescent by instilling a solution containing a preparation of a compound of formulae (I) to (XVI) into the anterior chamber of the eye after lens removal.
  • the solution can be osmotically balanced to provide minimal effective dosage when instilled into the anterior chamber of the eye, thereby inhibiting subcapsular epithelial growth with some specificity.
  • the compounds of the invention can also be used in the treatment of corneopathies marked by corneal epithelial cell proliferation, as for example in ocular epithelial disorders such as epithelial downgrowth or squamous cell carcinomas of the ocular surface.
  • Hair is basically composed of keratin, a tough and insoluble protein; its chief strength lies in its disulphide bond of cystine.
  • Each individual hair comprises a cylindrical shaft and a root, and is contained in a follicle, a flask-like depression in the skin.
  • the bottom of the follicle contains a finger-like projection termed the papilla, which consists of connective tissue from which hair grows, and through which blood vessels supply the cells with nourishment.
  • the shaft is the part that extends outwards from the skin surface, whilst the root has been described as the buried part of the hair.
  • the base of the root expands into the hair bulb, which rests upon the papilla.
  • Cells from which the hair is produced grow in the bulb of the follicle; they are extruded in the form of fibers as the cells proliferate in the follicle.
  • Hair “growth” refers to the formation and elongation of the hair fiber by the dividing cells.
  • the common hair cycle is divided into three stages: anagen, catagen and telogen.
  • anagen the epidermal stem cells of the dermal papilla divide rapidly.
  • Daughter cells move upward and differentiate to form the concentric layers of the hair itself.
  • the transitional stage, catagen is marked by the cessation of mitosis of the stem cells in the follicle.
  • the resting stage is known as telogen, where the hair is retained within the scalp for several weeks before an emerging new hair developing below it dislodges the telogen-phase shaft from its follicle. From this model it has become clear that the larger the pool of dividing stem cells that differentiate into hair cells, the more hair growth occurs.
  • the compounds of the invention can be employed as a way of reducing the growth of human hair as opposed to its conventional removal by cutting, shaving, or depilation.
  • the present method can be used in the treatment of trichosis characterized by abnormally rapid or dense growth of hair, e.g. hypertrichosis.
  • subject compounds can be used to manage hirsutism, a disorder marked by abnormal hairiness.
  • the compounds of the invention can also provide a process for extending the duration of depilation.
  • a subject compound will often be cytostatic to epithelial; cells, rather than cytotoxic, such agents can be used to protect hair follicle cells from cytotoxic agents which require progression into S-phase of the cell-cycle for efficacy, e.g. radiation-induced death.
  • Treatment by the compounds of the invention can provide protection by causing the hair follicle cells to become quiescent, e.g., by inhibiting the cells from entering S phase, and thereby preventing the follicle cells from undergoing mitotic catastrophe or programmed cell death.
  • compounds of the invention can be used for patients undergoing chemo- or radiation-therapies which ordinarily result in hair loss.
  • the subject treatment can protect hair follicle cells from death which might otherwise result from activation of cell death programs.
  • the administration of compounds of the invention can be stopped with concommitant relief of the inhibition of follicle cell proliferation.
  • the compounds of the invention can also be used in the treatment of folliculitis, such as folliculitis decalvans, folliculitis ulerythematosa reticulata or keloid folliculitis.
  • folliculitis such as folliculitis decalvans, folliculitis ulerythematosa reticulata or keloid folliculitis.
  • a cosmetic preparation of a compound of formulae (I) to (XVI) can be applied topically in the treatment of pseudofolliculitis, a chronic disorder occurring most often in the submandibular region of the neck and associated with shaving, the characteristic lesions of which are erythematous papules and pustules containing buried hairs.
  • the compounds of the invention can be used to induce differentiation and/or inhibit proliferation of epithelially derived tissue.
  • Such forms of these molecules can provide a basis for differentiation therapy for the treatment of hyperplastic and/or neoplastic conditions involving epithelial tissue.
  • preparations can be used for the treatment of cutaneous diseases in which there is abnormal proliferation or growth of cells of the skin.
  • the pharmaceutical preparations of the invention are intended for the treatment of hyperplastic epidermal conditions, such as keratosis, as well as for the treatment of neoplastic epidermal conditions such as those characterized by a high proliferation rate for various skin cancers, as for example basal cell carcinoma or squamous cell carcinoma.
  • the compounds of the invention can also be used in the treatment of autoimmune diseases affecting the skin, in particular, of dermatological diseases involving morbid proliferation and/or keratinization of the epidermis, as for example, caused by psoriasis or atopic dermatosis.
  • psoriasis squamous cell carcinoma
  • keratoacanthoma actinic keratosis
  • psoriasis which is characterized by scaly, red, elevated plaques on the skin
  • the keratinocytes are known to proliferate much more rapidly than normal and to differentiate less completely.
  • the preparations of the present invention are suitable for the treatment of dermatological ailments linked to keratinization disorders causing abnormal proliferation of skin cells, which disorders may be marked by either inflammatory or noninflammatory components.
  • therapeutic preparations of an inventive compound e.g., which promotes quiescense or differentiation, can be used to treat varying forms of psoriasis, be they cutaneous, mucosal orungual.
  • Psoriasis as described above, is typically characterized by epidermal keratinocytes which display marked proliferative activation and differentiation along a "regenerative" pathway.
  • Treatment with an antiproliferative compound of the invention can be used to reverse the pathological epidermal activiation and can provide a basis for sustained remission of the disease.
  • keratotic lesions are also candidates for treatment with the compounds of the invention.
  • Actinic keratoses for example, are superficial inflammatory premalig-nant tumors arising on sun-exposed and irradiated skin. The lesions are erythematous to brown with variable scaling.
  • Current therapies include excisional and cryosurgery. These treatments are painful, however, and often produce cosmetically unacceptable scarring.
  • treatment of keratosis such as actinic keratosis, can include application, preferably topical, of a composition containing at least one compound of formulae (I) to (XVI) in amounts sufficient to inhibit hyperproliferation of epidermal/epidermoid cells of the lesion.
  • Acne represents yet another dermatologic ailment which may be treated by the compounds of the invention.
  • Acne vulgaris for instance, is a multifactorial disease most commonly occurring in teenagers and young adults, and is characterized by the appearance of inflammatory and noninflammatory lesions on the face and upper trunk.
  • the basic defect which gives rise to acne vulgaris is hypercornification of the duct of a hyperactive sebaceous gland. Hypercornification blocks the normal mobility of skin and follicle microorganisms, and in so doing, stimulates the release of lipases by Propinobac-terium acnes and Staphylococcus epidermidis bacteria and Pitrosporum ovale, a yeast.
  • Treatment with an antiproliferative compound of formulae (I) to (XVI) may be useful for preventing the transitional features of the ducts, e.g. hypercornification, which lead to lesion formation.
  • the subject treatment may further include, for example, antibiotics, retinoids and antiandrogens.
  • the present invention also provides a method for treating various forms of dermatitis.
  • Dermatitis is a descriptive term referring to poorly demarcated lesions which are either pruritic, erythematous, scaly, blistered, weeping, fissured or crusted. These lesions arise from any of a wide variety of causes.
  • the most common types of dermatitis are atopic, contact and diaper dermatitis.
  • seborrheic dermatitis is a chronic, usually pruritic, dermatitis with erythema, dry, moist, or greasy scaling, and yellow crusted patches on various areas, especially the scalp, with exfoliation of an excessive amount of dry scales.
  • the compounds of the invention can also be used in the treatment of stasis dermatitis, an often chronic, usually eczematous dermatitis.
  • Actinic dermatitis is dermatitis that due to exposure to actinic radiation such as that from the sun, ultraviolet waves or x-or gamma-radiation.
  • the compounds of the invention can be used in the treatment and/or prevention of certain symptoms of dermatitis caused by unwanted proliferation of epithelial cells.
  • Such therapies for these various forms of dermatitis can also include topical and systemic corticosteroids, antipuritics, and antibiotics.
  • Ailments which may be treated by the compounds of the invention are disorders specific to non-humans, such as mange.
  • the compounds of the invention can be used in the treatment of human cancers, particularly basal cell carcinomas and other tumors of epithelial tissues such as the skin.
  • compounds of the invention can be employed, in the subject method, as part of a treatment for basal cell nevus syndrome (BCNS), and other other human carcinomas, adenocarcinomas, sarcomas and the like.
  • BCNS basal cell nevus syndrome
  • the compounds of the invention are used as part of a treatment of prophylaxis regimen for treating (or preventing) basal cell carcinoma.
  • the deregulation of the Wnt signaling pathway may be a general feature of basal cell carcinomas caused by ptc mutations. Consistent overexpres-sion of human ptc mRNA has been described in tumors of familial and sporadic BCCs, determined by in situ hybridization. Mutations that inactivate ptc may be expected to result in overexpression of mutant Ptc, because ptc displays negative autoregulation. Likewise, mutations that inactivate Wnt may be expected to result in overexpression of mutant Wnt, because Wnt displays negative autoregulation.
  • the compounds of the invention can also be used to treat patients with BCNS, e.g., to prevent BCC or other effects of the disease which may be the result of Wnt-mediated disorders.
  • Basal cell nevus syndrome is a rare autosomal dominant disorder characterized by multiple BCCs that appear at a young age.
  • BCNS patients are very susceptible to the development of these tumors; in the second decade of life, large numbers appear, mainly on sun-exposed areas of the skin. This disease also causes a number of developmental abnormalities, including rib, head and face alterations, and sometimes Polydactyly, syndactyly, and spina bifida.
  • the present invention provides pharmaceutical preparations and methods for controlling the formation of megakaryocyte-derived cells and/or controlling the functional performance of megakaryocyte-derived cells. For instance, certain of the compositions disclosed herein may be applied to the treatment or prevention of a variety hyperplastic or neoplastic conditions affecting platelets.
  • the invention relates to pharmaceutical compositions comprising at least one compound according to the invention.
  • the invention relates to pharmaceutical compositions comprising at least one compound according to the invention in a mixture with an inert carrier, where said inert carrier is a pharmaceutical carrier.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the compound is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, including but not limited to peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered orally.
  • Saline and aqueous dextrose are preferred carriers when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions are preferably employed as liquid carriers for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences” by E.W. Martin. Such compositions will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.
  • the invention further relates to a process for the preparation of a medicament comprising the steps of: a) preparing at least one compound according to the formulae (I) to (XVI); and b) formulating a medicament containing at least said compound; and to a method of treating a mammal for modulating the Wnt signalling pathway wherein the method comprises administering to said mammal a therapeutically effective amount of a compound according to formulae (I) to (XVI).
  • the compounds according to the invention used for the preparation of a medicament for the modulation of the Wnt signalling pathway in a mammal may be administered in any convenient route.
  • the compounds are formulated to be compatible with the desired route of administration and may be administered together with other biologically active agents.
  • the compounds may be formulated for the intravenous, intradermal, subcutanus, intramuscular, intraperitoneal, epidural, oral, transdermal, transmucosal, rectal or pulmonary administration. Administration can be systemic or local. Pulmonary administration can be employed by use of an inhaler or nebulizer, and formulation with an aerosolizing agent, for example.
  • the compound can be delivered in a vesicle, in particular a liposome (Langer (1990) Science 249, 1527.
  • the compound can be delivered via a controlled release system.
  • a pump may be used (Sefton (1987) CRC Crit. Ref. Biomed. Eng. 14, 201; Buchwald et al.
  • a controlled release system can be placed in proximity of the therapeutic target, i.e., the brain, thus requiring only a fraction of the systemic dose (e.g. Goodson, 1984, In: Medical Applications of Controlled Release, supra, Vol. 2, 115). Other controlled release systems are discussed in the review by Langer (1990, Science 249, 1527).
  • Toxicity and therapeutic efficacy of the compounds of the invention can be determined by standard pharmaceutical procedures in cell cultures or experimental animals.
  • the results obtained from the cell culture assays and animal studies can be used in formulating the range of dosage for use in medicaments in humans.
  • the specific dosage for any particular subject is influenced by several factors, e.g. by the activity of the specific compound used, the age, body weight, general health, gender and diet of the subject, the time and the route of administration and the rate of excretion.
  • 3-Aminopyarzole-4-carboxylic acid (5.00 g, 39.3 mmol), tolualdehyde (5.64 g, 47 mmol) and dimedone (6.60 g, 47 mmol) were taken in 2-propanol (50 ml) in a 250 ml RB flask equipped with a magnetic stirrer and refluxed with stirring under LC-MS control for 4 h whereby the reaction was complete.
  • reaction mixture was cooled down to room temperature, diluted with cyclohexane (100 ml), stirred for 1 h at room temperature, solids were collected by filtration, washed first with 1: 1 mixture of cyclohexane-diethylether (50 ml) and then with ice cold water and dried under high vacuum to yield 10.8 g (30.7 mmol, 78.1%) of the pure white solids of the title compound.
  • 3-Aminopyarzole-4-carboxylic acid ethyl ester (31.2 g, 200 mmol), tolualdehyde (26 ml, 220 mmol), dimedone (31 g, 220 mol) and acetic acid (5 ml) were taken in toluene (250 ml) in a 500 ml RB flask equipped with a magnetic stirrer and refluxed with stirring under LC-MS control for 8h whereby the reaction was complete.
  • reaction mixture was cooled down to room temperature, diluted with cyclohexane (250 ml), stirred for 2 h, solids were collected by filtration, washed first with 1: 1 mixture of cyclohexane-diethylether (50 ml) and then with 40% aqueous 2-propanol (100 ml) and dried under high to yield 66.4 g (175 mmol, 87.6%) of the pure white solids of the title compound.
  • reaction mixture was cooled down to room temperature, excess potassium carbonate was neutralized with acetic, partitioned between DCM (100 ml) and water (100 ml), organic phase was separated, dried over magnesium sulfate, solvent was removed under vacuum a 40 °C and then dried under high vacuum to yield 9.6 g (24.4 mmol, 97.7%) of the title compound as pale yellow solid.
  • reaction mixture was diluted with ice cold water (500 ml), solids were collected by filtration and washed with water to get light yellow crystals of the title compound (15.8 g, 47.8 mmol, 86.8%).
  • reaction mixture was cooled down to room temperature, excess potassium carbonate was neutralized with acetic, partitioned between DCM (100 ml) and water (100 ml), organic phase was separated, dried over magnesium sulfate, solvent was removed under vacuum a 40 °C and then dried under high vacuum to yield 11.5 g (27.5 mmol, 91.6%) of the title compound as white solid.
  • reaction mixture was diluted with water to 250 ml, stored at -20C for 2h, solids were collected by filtration, washed with ice cold water and then dried to yield 7.4g (21.2 mmol, 73.3%) of the pure light yellow solids of the title compound.
  • reaction mixture was cooled down to room temperature. It was then partitioned between DCM (200 ml) and water (200 ml), the organic phase was separated, dried over magnesium sulfate and the solvent was evaporated under vacuum at 40 °C to dryness giving 22g (55.6 mmol, 83.7%) of the light yellowish solids of the title product.
  • reaction mixture was diluted with 1: 1 mixture of DMSO/CAN to 2 ml, filter through a microfilter and the product was purified by HPLC to yield 44 mg of the pure product.
  • a reporter gene based assay describing the modulation of the TCF4 transcription factor was used. More specifically 4000 Hek293T cells were seeded into 384 high density plates. 24h after seeding a Wnt-sensitive reporter (6xTCF-luciferase (Firefly) (pTOP-FLASH; "Armadillo coactivates transcription driven by the product of the Drosophila segment polarity gene dTCF", Cell, 1997, 88(6), pages 789-99)) and constitutively expressed control reporter (Renilla luciferase pCMV-RL) were transfected into Hek293T.
  • a Wnt-sensitive reporter (6xTCF-luciferase (Firefly)
  • pTOP-FLASH "Armadillo coactivates transcription driven by the product of the Drosophila segment polarity gene dTCF", Cell, 1997, 88(6), pages 789-99
  • constitutively expressed control reporter Renilla lucifer
  • Wnt signaling was stimulated by cotransfecting mouse Wntl, mouse Frizzled 8 and human LRP6 according to "Casein kinase 1 gamma couples Wnt receptor activation to cytoplasmic signal transduction", Nature, 2005, 438(7069), pages 867-872.
  • 24 h after pathway stimulation compounds were added at a concentration of 10 microM and allowed to incubate for 24 h.
  • the respective compound was applied in increasing concentrations yielding final concentrations per well of 5nM - 100 microM.
  • cytotoxicity 24h after compound addition cytotoxicity was evaluated.
  • the media was removed and CellTiterGlo was added according to the manufactors manual.
  • the luciferase emission readout is directly correlated with the cellular amount of ATP, low luciferase emission thus is reflecting cytotoxicity of a compound.
  • the compound was applied in increasing concentrations yielding final concentrations per well of 5nM - 100 microM.
  • IC 5 o-values of the cytotoxic activity of compounds (23) to (28) against Hek293T and HepG2 are shown in table 2.
  • Table 2 IC 50 -values against Hek293T and HepG2 of compounds (23) to (28)
  • HCT116 human colorectal cancer cells
  • SW480 denoted (2)
  • Dld-1 denoted (3)
  • human fibroblasts HCT68, denoted (4)
  • Mc Coy's (1)
  • Dulbecco's Modified Eagles Medium (2)-(4)
  • fetal calf serum 1% penicillin/streptomycin
  • Cells were grown in T75 flasks at 37 °C, 5% C02 and trypsinized when 60-80% confluent by adding 2 ml of 0.25% Trypsin-EDTA (ethylenediaminetetraacetic acid) solution. Cells were then re-suspended into culture medium yielding approx. 750 cells, suspended in 45 microl medium, were plated into each well of a black 384-well plate for fluorescence imaging experiments. 24 - 36 hr later 5 microl compound solution (100 microM compound dissolved in ultra pure water containing 1% DMSO (dimethylsulf oxide)), were added to achieve a final concentration of 10 microM and were incubated for at least 72 hr.
  • Trypsin-EDTA ethylenediaminetetraacetic acid

Abstract

Compounds according to the general Formula (I), wherein X1 is CR1 or N; X2 is CR2 or N; X3 is CR3 or N; with at least one of the group X1; X2; and X3 is N; Z is CR4 or CR4R5; Y is N or NR6; the dotted line denotes 1 or 2 double bonds; L is O or NR7; each A is independently from each other CR8 R 8a which is same or different and one A is optionally selected from the group consisting of O; S; NR9; S(O) and S(0)2; n is 2 to 4; their solvates, hydrates, esters and pharmaceutically acceptable salts, their use for modulating the Wnt signalling pathway activity and their use as a medicament, preferably for the treatment of cancer.

Description

Pyrazol[l,5-a]pyrimidine derivatives as Wnt pathway antagonists
The present invention relates to compounds having the general formula (I) with the
1 3
definitions of X - X Y, Z, L, A and n given below and/or solvates, hydrates, esters and pharmaceutically acceptable salts thereof. Furthermore, the invention relates to the use of said compounds for modulating of the Wnt signalling pathway activity and their use as a medicament, preferably for the treatment of cancer.
The Wnt signalling pathway plays an important role in the regulation of cell proliferation and differentiation. Aberrant activation of the Wnt signalling pathway is known to promote uncontrolled cell growth and survival and can therefore be a major driving force in a broad spectrum of human cancers and diseases. For example, the inhibition of aberrant Wnt signalling pathway activity in cancer cell lines effectively blocks their growth (N. Barker and H. Clevers "Mining the Wnt pathway for cancer therapeutics", Nature Reviews, vol. 5, 2007, pages 997-1014; R. Nusse, "Wnt signalling in disease and in development", Cell Research, Vol. 15, 2005, pages 23-32). Other disorders and diseases are considered to be influenced by an aberrant Wnt signalling pathway, too (see e.g. literature cited above).
The Wnt signalling pathway involves a large number of proteins regulating the production of Wnt signalling molecules, their interaction with receptors on target cells and the physiological response of target cells resulting from the exposure of cells to the extra- cellular Wnt ligands.
Secreted signalling proteins of the Wnt family bind to specific Frizzled (Frz) receptor complexes on the surface of target cells and activate distinct intracellular pathways that are broadly classified as canonical or non-canonical Wnt signalling pathways.
In brief, the canonical pathway regulates the amount of the protein beta-catenin in a cell and its ability to enter the nucleus of the cell, where it interacts with members of the Tcf/Lef protein family. Beta-catenin and Tcf form active transcription factor complexes in the nucleus and activate the Wnt target gene. The presence of the Tcf-beta-catenin complex in the nucleus is a hallmark in the Wnt signalling pathway indicating its activation. An overview of the Wnt signalling pathway can be found in N. Barker and H. Clevers "Mining the Wnt pathway for cancer therapeutics", Nature Reviews, vol. 5, 2007, pages 997-1014.
The presence of Tcf-beta-catenin complexes in the nuclei of cells leads to activation of the genetic program considered to promote cancer formation by stimulating cell growth, blocking apoptosis and altering cell movement. For instance, the artificial disruption of Tcf-beta-catenin complex formation in colon cancer cells effectively blocks target gene activation and inhibits the growth in vitro. Drugs designed to inhibit the Wnt signalling pathway and consequently the formation of the Tcf-beta-catenin complex in the nucleus of a cell are therefore expected to hold great potential for the treatment of a range of cancers and other diseases associated with the Wnt signalling pathway.
Therefore, there is a strong need for novel compounds which modulate the Wnt signalling pathway thereby opening new routes for the treatment of disorders and/or diseases associated with an aberrant activation of Wnt signalling.
An object of the present invention is to provide such compounds. This object is achieved by a compound having the general formula (I)
Figure imgf000003_0001
wherein
X1 is CR1 or N:
X2 is CR2 or N:
X3 is CR3 or N:
wherein at least one of the group % and X 3
Figure imgf000003_0002
X2 J is N;
Z is CR4 or CR4R5;
Y is N or NR6;
the dotted line denotes 1 or 2 double bonds;
L is O or NR7; each A is independently from each other CR 8 R 8ίΐ which is same or different and one A is optionally selected from the group consisting of O; S; NR9; S(O) and S(0)2;
n is 2 to 4;
R 1 ; R2"; and R 3J are independently from each other selected from H; OH; halogen;
CN; Ci-C6 alkyl; C3-C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl; C4-Q5 aralkyl; C4-Q5 heteroarylalkyl; ORla" C(0)Rla; C(0)ORla; C(0)NRlaRlb; C(0)N(Rla)ORlb C(0)N(Rla)N(Rlb)C(0)Rlc; C(0)N(Rla)N(Rlb)Rlc; S(0)2N(RlaRlb):
S(0)N(RlaRlb); S(0)2Rla; S(0)Rla; N(Rla)S(0)2N(RlbRlc); SRla; OC(0)Rla N(Rla)C(0)Rlb; N(Rla)S(0)2Rlb; N(Rla)S(0)Rlb; N(Rla)C(0)N(RlbRlc): N(Rla)C(0)ORlb; OC(0)N(RlaRlb); CHN(Rla); and CHNN(RlaRlb) wherein alkyl; cycloalkyl; heterocyclyl; aryl; heteroaryl; aralkyl; and heteroarylalkyl are optionally substituted by one ore more groups R10 which are same or different;
Rla; Rlb; and Rlc are independently from each other selected from H; C C6 alkyl; C3- C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl; C8-C12 biaryl; C8-C12 heterobiaryl; C4-Q5 aralkyl; and C4-Q5 heteroarylalkyl; wherein alkyl; cycloalkyl; heterocyclyl; alkenyl; aryl; biaryl; heteroaryl; heterobiaryl; aralkyl; and heteroarylalkyl are optionally substituted with one or more R10 which are the same or different ; R10 is selected from halogen, CN, OH, C1-6 alkyl; C3-C7 cyclyl; C3-C7
heterocyclyl; C3-C7 aryl; C3-C7 heteroaryl; C8-C12 biaryl; C8-C12
heterobiaryl; OR10a; C(O)R10a; C(O)OR10a; NR10aR10b; C(O)NR10aR10b;
S(O)2N(R10aR10b); S(O)N(R10aR10b); S(O)2R10a; S(O)R10a;
N(R10a)S(O)2N(R10bR10c); SR10a; OC(O)R10b; N(R10a)C(O)R10b;
N(R10a)S(O)2R10b; N(R10a)S(O)R10b; N(R10a)C(O)N(R10bR10c);
N(R10a)C(O)OR10b; and OC(O)N(R10aR10b); wherein cyclyl; heterocyclyl; aryl; heteroaryl; biaryl; heterobiaryl; and alkyl are optionally substituted with one or more R10a, which are the same or different; R10a; R10b and R10c are independently from each other selected from H; halogen; C3-
C7 cyclyl; C3-C7 heterocyclyl; C3-C7 aryl; C3-C7 heteroaryl; and C C6 alkyl; wherein cyclyl; heterocyclyl; aryl; heteroaryl and alkyl are optionally substituted with one or more halogen which are the same or different;
R4 is a 5 to 6 membered aromatic or heteroaromatic ring which is optionally substituted by one or more R4a; selected from halogen, CN, OH, OR , C(0)R4B; C(0)0R4B; NR R C(0)NR4bR4c; S(0)2N(R4bR4c); S(0)N(R4bR4c); S(0)2R4b; S(0)R4b N(R4b)S(0)2N(R4cR4d); SR4b; OC(0)R4b; N(R4b)C(0)R4c; N(R4b)S(0)2R4c N(R4b)S(0)R4c; N(R4b)C(0)N(R4cR4d); N(R4b)C(0)OR4c; OC(0)N(R4b R4c) and CrC6 alkyl; wherein alkyl is optionally substituted with one or more R1 which are the same or different ;
R1J is selected from halogen, CN, OH, C1-6 alkyl; OR , C(0)R1Ja; C(0)OR1Ja;
NRi3aRi3b. C(0)NR13aR13b; S(0)2N(R13aR13b); S(0)N(R13aR13b); S(0)2R13a; S(0)R13a; N(R13a)S(0)2N(R13bR13c); SR13a; OC(0)R13a; N(R13a)C(0)R13b; N(R13a)S(0)2R13b; N(R13a)S(0)R13b; N(R13a)C(0)N(R13bR13c);
N(R13a)C(0)OR13b; and OC(0)N(R13a R13b); wherein C1-6 alkyl is optionally substituted with one or more halogen, which are the same or different;
R4b, R4c, R4d, R13a; R13b and R13c are independently from each other selected from H; and CrC6 alkyl; wherein C1-6 alkyl is optionally substituted with one or more halogen, which are the same or different;
R5 is H or linear CrC6 alkyl;
R6 is selected from H; OH; CrC6 alkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl;C4-C15 aralkyl; C4-C15 heteroarylalkyl; OR6a; C(0)R6a; C(0)OR6a; OC(0)R6a; OC(0)N(R6aR6b); N(R6aR6b); N(R6a)C(0)N(R6bR6c); and N(R6a)C(0)OR6b wherein alkyl; cycloalkyl; heterocyclyl; alkenyl; aryl; heteroaryl; aralkyl; and heteroarylalkyl are optionally substituted by one ore more R11, which are the same or different;
R6a; R6b; and R6c are independently from each other selected from H; C C6 alkyl; C3- C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl; C4-C15 aralkyl; and C4-C15 heteroarylalkyl which are optionally substituted with one or more R11, which are the same or different;
R11 is selected from C1-6 alkyl; halogen, CN; OH; ORlla; C(0)Rlla; C(0)ORlla NRiiaRi ib. C(0)NRllaRllb; S(0)2N(RllaRllb); S(0)N(RllaRllb); S(0)2Rlla S(0)Rlla; N(Rlla)S(0)2N(RllbRl lc); SRlla; OC(0)Rllb; N(Rlla)C(0)Rllb N(Rl la)S(0)2Rlla; N(Rl la)S(0)Rllb; N(Rlla)C(0)N(RllbRllc):
N(Rl la)C(0)ORl lb; and OC(0)N(RllaRllb); wherein C1-6 alkyl is optionally substituted with one or more halogen, which are the same or different;
Rlla; Rllb; and Rllc are independently from each other selected from H; and C C6 alkyl; wherein Ci_6 alkyl is optionally substituted with one or more halogen, which are the same or different; R7 is selected from H; OH; CrC6 alkyl; C3-C7 cycloalkyl; C3-C7 heterocyclyl; C2-
C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl; C4-C15 aralkyl; C4-C15 heteroarylalkyl; OR7a; C(0)R7a; C(0)OR7a; OC(0)R7a; OC(0)N(R7aR7b); N(R7aR7b); N(R7a)C(0)R7b; N(R7a)C(0)N(R7bR7c); and N(R7a)C(0)OR7b; wherein alkyl; cycloalkyl; alkenyl; aryl; aralkyl; and heteroarylalkyl are optionally substituted by one ore more R 12 , which are the same or different;
R a; R ; and R c are independently from each other selected from H; C C6 alkyl; C3- C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl; C4-Q5 aralkyl; and C4-Q5 heteroarylalkyl wherein alkyl; cycloalkyl; heterocyclyl; alkenyl; aryl; heteroaryl; aralkyl; heteroarylalkyl are optionally substituted with one or more R 12 , which are the same or different ;
R1Z is selected from C1-6 alkyl; halogen, CN, OH, OR1/a; C(0)R1/a; C(0)OR1/a NR12aR12b; C(0)NR12aR12b; S(0)2N(R12aR12b); S(0)N(R12aR12b); S(0)2R12a
S(0)R12a; N(R12a)S(0)2N(R12bR12c); SR12a; OC(0)R12b; N(R12a)C(0)R12b N(R12a)S(0)2R12a; N(R12a)S(0)R12b; N(R12a)C(0)N(R12bR12c):
N(R12a)C(0)OR12b; and OC(0)N(R12aR12b); wherein C1-6 alkyl is optionally substituted with one or more halogen, which are the same or different; R a; R and R c are independently from each other selected from H; and CrC6 alkyl; wherein C1-6 alkyl is optionally substituted with one or more halogen; which are the same or different; R 8 ; and R 8<i are independently from each other selected from H and C¾;
R9 is selected from OH; CrC6 alkyl; C3-C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl; C4-Q5 aralkyl; C4-Q5 heteroarylalkyl; OR9a; C(0)R9a and C(0)OR9a; wherein alkyl; cycloalkyl; alkenyl; aryl; aralkyl; and heteroarylalkyl are optionally substituted by one ore more groups selected from OH; halogen; CN; CrC6 alkyl; OR9b;
C(0)R9b; C(0)OR9b; NR9bR9c; C(0)NR9bR9c; S(0)2N(R9bR9c);
S(0)N(R9bR9c); S(0)2R9b; S(O) R9b; N(R9b)S(0)2N(R9cR9d); SR9b;
OC(0)R9b; N(R9b)C(0)R9c; N(R9b)S(0)2R9c; N(R9b)S(0)R9c; N(R9b)C(0)N(R9cR9d); N(R9b)C(0)OR9c; and OC(0)N(R9bR9c); wherein C1-6 alkyl is optionally substituted with one or more halogen, which are the same or different;
R9a; R9b and R9c are independently from each other selected from H; and CrC6 alkyl; wherein C1-6 alkyl is optionally substituted with one or more halogen; which are the same or different; and/or solvates; hydrates; esters; and pharmaceutically acceptable salts thereof.
"Alkyl" means a linear or branched saturated aliphatic hydrocarbon group e.g. methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, n-pentyl, i-pentyl, n-hexyl, i-hexyl and 3-methyl- pentyl and the like.
"Alkenyl" means a linear or branched unsaturated aliphatic hydrocarbon group with at least one double bond e.g. ethenyl, propenyl (allyl), 1-butenyl, 2-butenyl, 1-pentenyl, 2- pentenyl, 1-hexenyl, 2-hexenyl and 3-hexenyl and the like.
"Cycloalkyl" means a saturated 3 to 8-member hydrocarbon ring e.g. cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. "Heterocyclyl" means a saturated 3- to 8-member hydrocarbon ring containing 1 to 4 heteroatoms selected from the group N, O and S in the ring. The one or more heteroatoms present in the ring replace a -CH2- group in the ring. Preferred C3-C8 heterocyclyl groups containing 1 to 4 heteroatoms selected from the group N, O and S are derived from the following heterocyclic compounds: tetrahydrofurane, pyrrolidine, tetrahydrothiophene, oxazolidine, piperidine, tetrahydropyrane, piperazine, dioxane, morpholine and trioxane.
"Aryl" means an aromatic 3- to 7-numbered hydrocarbon ring e.g. phenyl and the like.
"Heteroaryl" means an aromatic 3- to 7-numbered hydrocarbon ring containing 1 to 4 heteroatoms selected from the group N, O and S in the ring. The one or more heteroatoms present in the ring replace a -CH= group in the ring. Preferred C3-C7 heteroaryl groups containing 1 to 4 heteroatoms selected from the group N, O and S are derived from the following hetero aromatic compounds: Pyrrole, pyrazole, imidazole, triazole, tetrazole, furane, thiophene, oxazole, isoxazole, oxadiazole, thiazole, isothiazole, thiadiazole, pyridine, pyridazine, pyrimidine, pyrazine and triazine.
"Aralkyl" means a C3-C7 aryl group substituted with at least one Ci-Cg alkyl group, wherein aryl and alkyl are defined as above. The aralkyl group may be bound via the Ci-Cg alkyl group or via the C3-C7 aryl group. "Heteroarylalkyl" means a C3-C7 heteroaryl group substituted with at least one Ci-Cg alkyl group, wherein heteroaryl and alkyl are defined as above. The heteroarylalkyl group may be bound via the Ci-Cg alkyl group or via the C3-C7 heteroaryl group.
"Biaryl" means an aromatic hydrocarbon with two condensed rings with 8 to 12 carbon atoms. C8-C12 biaryl groups are for example derived from naphthalene and indene.
"Heterobiaryl" means an aromatic 8- to 12-numbered hydrocarbon with two condensed rings containing in total 1 to 4 heteroatoms selected from the group N, O and S in the ring. The one or more heteroatoms present in the ring replace a -CH= group in the ring. C8-C12 heterobiaryl groups containing 1 to 4 heteroatoms selected from the group N, O and S are for example derived from the following heteroaromatic compounds: indole, benzofurane, benzimidazole, purine, benzothiophen, benzothiazole, isoquinoline, quinoxaline, quinoline and quinazoline, In a preferred embodiment of the invention at least one of the group X 11, X2" and X 3J is N, i.e. at least X3 is N (Ila), at least X2 is N (lib) or at least X1 is N (lie). Subsequently the compound has one of the following formulae (Ila) to (lie):
Figure imgf000009_0001
(Ila) (lib) (lie) wherein the definitions of the substituents given above.
Further preference is given to an embodiment of the invention wherein at least two of the group X2" and X 3J are N, giving the compounds according to formulae (Ilia) to (IIIc):
Figure imgf000009_0002
(Ilia) (nib) (IIIc) wherein the definitions of the substituents given above.
In another preferred embodiment of the invention X 1 , X2 and X 3 are N giving the compound of the following formula (IV):
Figure imgf000009_0003
wherein the substituents are defined as above.
According to a further preferred embodiment of the invention Y is NR6, Z is CR4R5 and at least one of the group X 1 , X2 and X 3 is N and the compound has the formula (V):
Figure imgf000010_0001
wherein the substituents are defined as above.
According to a further preferred embodiment of the invention Y is NR6, Z is CR4R5 and at least X 3 (Via), at least X 2 (VIb), or at least X 1 (Vic) is N. The compound according to this embodiment has one of the formulae (Via) to (Vic):
Figure imgf000010_0002
(Via) (VIb) wherein the substituents are defined as above.
According to another preferred embodiment of the invention Y is NR6, Z is CR4R5 and at least X2 and X3 are N, at least X1 and X3 are N or at least X1 and X2 are N. In this embodiment the compound has one of the formulae (Vila) to (VIIc):
Figure imgf000010_0003
(Vila) (Vllb) (VIIc) wherein the substituents are defined as above. Preference is also given to an embodiment of the invention wherein Y is NR6, Z is CR4R5 and X 11, X2" and X 3J are N. In this embodiment of the invention the compound has the formula (VIII):
Figure imgf000011_0001
wherein the substituents are defined as above.
In another preferred embodiment of the invention Y is N and the compound has the formula (IX):
Figure imgf000011_0002
wherein the substituents are defined as above.
According to a further preferred embodiment of the invention Y is N and at least X is N
(Xa), at least X 2 is N (Xb) or at least X 1 is N (Xc). This leads to the compounds of formulae (Xa) to (Xc):
Figure imgf000011_0003
(Xa) (Xb) (Xc) wherein the substituents are defined as above. According to a another preferred embodiment of the invention Y is N, at least one of the group X1, X2 and X3 is N and Z is CR4R5 (XIa) or CR4 (Xlb). The compound of this embodiment has the formula (XIa) or (Xlb):
Figure imgf000012_0001
(XIa) wherein the substituents are defined as above.
Further preference is given to an embodiment of the invention wherein Y is N, Z is CR4R5 and at least X3 is N (Xal), at least X2 is N (Xbl) or at least X1 is N (Xcl) is N or Z is CR4 and at least X3 is N (Xa2), at least X2 is N (Xb2) or at least X1 is N (Xc2) is N . The compound of this embodiment has one of the formulae (Xal) to (Xc2):
Figure imgf000012_0002
wherein the substituents are defined as above.
Another preferred embodiment of the invention refers to the compound of the formulae (Xlla) to (XIIc) wherein Y is N and at least X2 and X3 are N (Xlla), at least X1 and X3 are N (Xllb) or at least X1 and X2 are N (XIIc):
Figure imgf000013_0001
(Xlla) (Xllb) (XIIc) wherein the substituents are defined as above.
According to a further preferred embodiment of the invention Y is N, Z is CR4 and at least X2 and X3 are N (Xllal), at least X1 and X3 are N (Xllbl) or at least X1 and X2 are N (XIIcl) or Z is CR4R5 and at least X2 and X3 are N (XIIa2), at least X1 and X3 are N
(XIIb2) or at least X 1 and X 2 are N (XIIc2). In this embodiment of the invention the compound has the formulae (Xllal) to (XIIc2):
Figure imgf000013_0002
wherein the substituents are defined as above.
Another preferred embodiment of the invention relates the compound wherein Y, X1, and X are N. In this embodiment of the invention compound has the formula (XIII):
Figure imgf000013_0003
(xiii), wherein the substituents are defined as above. According to a further preferred embodiment of the invention Y, X 1 , X2 and X 3 are N and Z is CR4 (XHIa) or CR4R5 (XHIb) and the compound has the formula (XHIa) or (XHIb): Formel XHIa ist in meiner Ansicht nicht vorhanden! !
Figure imgf000014_0001
wherein the substituents are defined as above.
Another preferred embodiment of the invention relates to the compound of formula (XIV) with n is 2:
Figure imgf000014_0002
wherein the substituents are defined as above.
According to a further preferred embodiment of the invention n is 3 and the compound has the formula (XV):
Figure imgf000014_0003
wherein the substituents are defined as above, and more preferred within this embodiment is
Figure imgf000014_0004
and even more preferred X is N. According to another preferred embodiment of the invention n is 4 and the compound has the formula (XVI):
Figure imgf000015_0001
wherein the substituents are defined as above.
Within the compounds of formulae (I) to (XVI) those compounds are especially prefered wherein
R4 is selected from C6 aryl and C6 heteroaryl and is substituted in p- and both m- positions by three substituents selected independently from each other from R4a as defined above, wherein the substituents of the two o-positions are preferably H. If Z is CR4R5 it is further preferred that R5 is H.
Within the compounds of formulae (I) to (XVI) those compounds are especially preferred too, wherein
X1 is CR1 with R1 selected from OH; halogen; Ci-C6 alkyl; C3-C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 heteroaryl; C4-C15 aralkyl; C4-C15
heteroarylalkyl; ORla; C(0)Rla; C(0)N(Rla)ORlb; C(0)N(Rla)N(Rlb)C(0)Rlc; C(0)N(Rla)N(Rlb)Rlc; S(0)2N(RlaRlb); S(0)N(RlaRlb); S(0)2Rla; S(0)Rla;
N(Rla)S(0)2N(RlbRlc); SRla; OC(0)Rla; N(Rla)S(0)2Rlb; N(Rla)S(0)Rlb;
N(Rla)C(0)N(RlbRlc); N(Rla)C(0)ORlb; OC(0)N(RlaRlb); CHN(Rla); and
CHNN(RlaRlb), in particular from C(0)N(Rla)ORlb; C(0)N(Rla)N(Rlb)Rlc; and C(0)N(Rla)N(Rlb)C(0)Rlc, and even more preferred R1 is selected from
C(0)N(H)ORlb; C(0)N(H)N(H)Rlc; and C(0)N(H)N(H)C(0)Rlc.
Within the compounds of formulae (I) to (XVI) those compounds are especially preferred
1 2 3 wherein at least one of XI, X2 and X3 is not N and the respective group R ; R or R is selected from C(0)N(Rla)ORlb; C(0)N(Rla)N(Rlb)Rlc; and C(0)N(Rla)N(Rlb)C(0)Rlc, and even more preferred R1; R2 or R3 is selected from C(0)N(H)ORlb; C(0)N(H)N(H)Rlc; and C(0)N(H)N(H)C(0)Rlc.
Particularly those compounds of formulae (I) to (XVI) are preferred, wherein R4 is selected from C6 aryl and C6 heteroaryl and is substituted in p- and both impositions by three substituents selected independently from each other from R4a as defined above, wherein the substituents of the two o-positions are preferably H and
X1 is CR1 with R1 selected from OH; halogen; Ci-C6 alkyl; C3-C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl; C4-C15 aralkyl; C4-C15 heteroarylalkyl; ORla; C(0)Rla; C(0)ORla; C(0)N(RlaRlb); C(0)N(Rla)ORlb; C(0)N(Rla)N(Rlb)C(0)Rlc; C(0)N(Rla)N(Rlb)Rlc; S(0)2N(RlaRlb);
S(0)N(RlaRlb); S(0)2Rla; S(0)Rla; N(Rla)S(0)2N(RlbRlc); SRla; OC(0)Rla;
N(Rla)C(0)Rlb; N(Rla)S(0)2Rlb; N(Rla)S(0)Rlb; N(Rla)C(0)N(RlbRlc);
N(Rla)C(0)ORlb; OC(0)N(RlaRlb); CHN(Rla); and CHNN(RlaRlb), in particular from C(0)N(Rla) ORlb; C(0)N(Rla)N(Rlb)Rlc; and C(0)N(Rla)N(Rlb)C(0)Rlc, and even more preferred from C(0)N(H)ORlb; C(0)N(H)N(H)Rlc; and
C(0)N(H)N(H)C(0)Rlc.
If Z is CR4R5 it is further preferred within the embodiment described above that R5 is H.
Within the compounds of formulae (I) to (XVI) those compounds are especially preferred wherein
R4 is selected from C6 aryl and C6 heteroaryl and is substituted in p- and both i positions by three substituents selected independently from each other from R4a as defined above, wherein the substituents of the two o-positions are preferably H and
1 2 3 wherein at least one of XI, X2 and X3 is not N and the respective group R ; R or R is selected from C(0)N(Rla)ORlb; C(0)N(Rla)N(Rlb)Rlc; and C(0)N(Rla)N(Rlb)C(0)Rlc, and even more preferred R1; R2 or R3 is selected from C(0)N(H)ORlb; C(0)N(H)N(H)Rlc; and C(0)N(H)N(H)C(0)Rlc.
If Z is CR4R5 it is further preferred within the embodiment described above that R5 is H.
According to the invention the solvates; hydrates; esters; and/or pharmaceutically acceptable salts of all compounds of formulae (I) to (XVI) described above are included herein.
Some of the compounds of the invention and/or salts or esters thereof will exist in different stereo isomeric forms. All of these forms are subjects of the invention.
Described below are exemplary salts of the compounds according to the invention which are included herein. The list of the different salts stated below is not meant to be complete and limiting. Compounds according to the invention which contain one or more acidic groups can be used according to the invention, e.g. as their alkali metal salts, alkaline earth metal salts or ammonium salts. More precise examples of such salts include sodium salts, potassium salts, calcium salts, magnesium salts or salts with ammonia or organic amines such as, e.g. ethylamine, ethanolamine, triethanolamine or amino acids.
Compounds according to the invention which contain one or more basic groups, i.e. groups which can be protonated, can be used according to the invention in the form of their addition salts with inorganic or organic acids.
Examples for suitable acids include hydrogen chloride, hydrogen bromide, phosphoric acid, sulfuric acid, nitric acid, methanesulfonic acid, p-toluenesulfonic acid, napthalenedisulfonic acid, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propionic acid, pivalic acid, diethylacetic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, malic acid, sulfamic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid and other acids known to a person skilled in the art.
Compounds according to the invention which contain several basic groups can simultaneously form different salts.
If a compound according to the invention simultaneously contains acidic and basic groups in the molecule, the invention also includes, in addition to the salt forms mentioned, inner salts or betaines.
The respective salts of the compounds according to the invention can be obtained by customary methods which are known to the person skilled in the art, 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 term "pharmaceutically acceptable" means approved by a regulatory agency such as the EMEA (Europe) and/or the FDA (US) and/or any other national regulatory agency for use in animals, preferably in humans. Furthermore, the invention includes all salts of the compounds according to the invention which, owing to low physiological compatibility, are not directly suitable for use in pharmaceuticals but which can be used, for example, as intermediates for chemical reactions or for the preparation of pharmaceutically acceptable salts or which might be suitable for studying Wnt signalling pathway modulating activity of a compound according of the invention in any suitable manner, such as any suitable in vitro assay.
The present invention furthermore includes all solvates of the compounds according to the invention.
The present invention furthermore includes derivatives/prodrugs (including the salts thereof) of the compounds according to the invention which contain physiologically tolerable and cleavable groups and which are metabolized in animals, preferably mammals, most preferably humans into a compound according to the invention.
The present invention furthermore includes the metabolites of the compounds according to the invention.
The term "metabolites" refers to all molecules derived from any of the compounds according to the invention in a cell or organism, preferably mammal.
Preferably the term "metabolites" relates to molecules which differ from any molecule which is present in any such cell or organism under physiological conditions. The structure of the metabolites of the compounds according to the invention will be obvious to any person skilled in the art, using the various appropriate methods.
The compounds of the invention may be prepared by the three component reaction of an aldehyde R4-CHO, wherein R4is defined as in formula (I) (e.g. benzaldehyde and substituted benzaldehydes, heteroaromatic aldehydes and substituted heteroaromatic aldehydes), a 5-to-7-membered cyclic 1,3-diketone (e.g. 1,3-cyclohexanone and its derivatives, 1,3-cylopentanone and its derivatives, 1,3-cycloheptanone and its derivatives, Thiopyran-3,5-dione and its derivatives, Pyran-3,5-dione and its derivatives, Piperidine- 3,5-dione and its derivatives, Piperidine-2,4-dione and its derivatives, Pyrrolidine-2,4- dione and its derivatives, Furan-2,4-dione and its derivatives), and an amino heteroaromate (e.g. 3-aminopyrazole and its derivatives, aminotriazole and its derivatives, aminoimidazole and its derivatives and aminotetrazole) in a solvent like toluene, alcohols or dimehylformamide. The reaction of benzaldehyde, 1,3-cyclohexanone and 3- aminopyrazole-4-carboxylic ethylester leading to compound i is shown as example in scheme I. Further modifications of compound i in one more steps deliver compounds like ii - viii which may serve for the synthesis of further compounds.
Figure imgf000019_0001
Scheme I
The invention further provides a process for the preparation of the inventive compounds of formulae (I) to (XVI) comprising reacting an aldehyde R4-CHO, a 5-to-7-membered cyclic 1,3-diketone and a heteroaromate substituted by NH2 wherein R4 is defined as in formula (I).
Depending on the circumstances of the individual case, in order to avoid side reactions during the synthesis of a compound of the general formulae (I) to (XVI), it can be necessary or advantageous to temporarily block functional groups by introducing protective groups and to deprotect them in a later stage of the synthesis, or to introduce functional groups in the form of precursor groups and at a later stage to convert them into the desired functional groups. Suitable synthetic strategies, protective groups and precursor groups are known to the person skilled in the art.
If desired, the compounds of the formulae (I) to (XVI) can be purified by customary purification procedures, for example by recrystallization or chromatography. The starting materials for the preparation of the compounds of the formulae (I) to (XVI) are commercially available or can be prepared according to or analogously to literature procedures.
The compounds of the invention may serve as a basis for the preparation of the other compounds according to the invention by several methods well known by the person skilled in the art.
The present invention relates to the discovery that signal transduction pathways regulated by Wnt can be inhibited, at least in part, by compounds of formulae (I) to (XVI). As set out in more detail below, these compounds can inhibit proliferation of tumor cells having Wnt modulated activity. Therefore, the compounds according to the invention are suited for modulating the Wnt signalling pathway.
As used herein, the term "modulating the Wnt signalling pathway" refers to an effect on the series of events that occur when Wnt proteins bind to cell- surface receptors of the frizzled family resulting in an accumulation of beta-catenin in the cell cytoplasm that reaches the nucleus of a cell, and consequently, the Wnt target genes are expressed. The Wnt signalling pathway may be modulated by direct or indirect modulation.
"Direct modulation" according to the present invention means an interaction of the inventive compounds with proteins directly involved in the Wnt signalling pathway leading to an increase or decrease of the expression of the Wnt target genes. "Indirect modulation" according to the present invention means an increase or decrease of the expression of the Wnt target genes without a direct interaction of the inventive compound with the components involved in the Wnt signalling pathway. Examples for the indirect modulation of the Wnt signalling pathway are tankyrase-inhibitors and calcium regulators like siperone, thapsigargine and iononycine.
Inhibition of tankyrases stabilizes one protein of the Wnt signalling pathway (axin), which, inhibits the Wnt signalling pathway. Inhibition of the decomposition of axin leads to an increase of axine and in turn to the inhibition of the Wnt signalling pathway (Huang et al., Nature 461, pp. 614 to 620 (2009))
By increasing the intracellular calcium level the Wnt protein beta-catenin is transferred out of the nucleus and decomposes. This inhibits a beta-catenin-mediated Wnt signalling pathway activity without inhibiting a Wnt protein directly, too ((Lu et al., BMC Pharm, 2009 9: 13 (doi: 10.1186/1471-2210-9-13) and Li et al., PNAS 99, pp. 13254 to 13259 (2002)).
While not wishing to be bound by any particular theory, the activation of a receptor may be the mechanism by which these compounds act as described in US 2007/0219257 Al. For example, the compounds could affect the activity of a Wnt frizzled receptor. Alternatively, the compounds could affect the activity of the serine/ threonine kinase GSK3B, which is involved in the down regulation of B-catenin. The compounds could also affect the activity of the APC gene. In the absence of Wnt signal, the APC protein functions to foster degradation of B-catenin and prevent its nuclear entry. Wnt stimulation, loss of APC protein function, or of its associated partner Axin, all lead to stabilization of and concentration in the nucleus of B-catenin, which then can act as a transcriptional co-activator by associating with the Tcf/LEF family of transcription factors. APC in complex with Axin and other proteins target B-catenin for proteasomal degradation by scaffolding the association between B-catenin and kinases whose action lead to B-catenin ubiquitinylation; this action is abrogated by recruitment of the degradation complex to the membrane upon Wnt activation of a receptor complex that includes Frizzled (Fz), a relative of Smo, and LRP5/6. The pathway can also be activated by mutations of B-catenin that render it resistant to degradation. Or, for example, the compounds could alter the activity of Dishevelled, which is a positive mediator of Wnt signaling. For example, the ability of these compounds to inhibit proliferation of cells may be due to the ability of such molecules to interact with Wnt, or at least to interfere with the ability of those proteins to activate a Wnt-mediated signal transduction pathway. Signal transduction antagonists of different structures, even ones that bind to the same protein in the signaling pathways, may act in slightly different ways. Accordingly, even if a particular condition caused or contributed to by aberrant or unwanted activation of the Wnt pathway shows little response to treatment by one of the antagonists disclosed herein, another of the antagonists disclosed herein may nonetheless be efficacious.
One embodiment of the present invention includes the use of compounds of formulae (I) to (XVI) that agonize inhibition of Wnt signaling, such as by inhibiting activation of Wnt downstream components of the signaling pathway, in the regulation of repair and/or functional performance of a wide range of cells, tissues and organs, including normal cells, tissues, and organs. For instance, the compounds of formulae (I) to (XVI) have therapeutic and cosmetic applications ranging from regulation of neural tissues, bone and cartilage formation and repair, regulation of spermatogenesis, regulation of smooth muscle, regulation of lung, liver and other organs arising from the primitive gut, regulation of hematopoietic function, regulation of skin and hair growth, etc. Moreover, the compounds of formula (I) can be applied to cells that are provided in culture (in vitro), or on cells in a whole animal (in vivo).
Another embodiment of present invention includes the use of compounds of formulae (I) to (XVI), which antagonize activity of the Wnt pathway resulting in the regulation of repair and/or functional performance of a wide range of cells, tissues, and organs. For instance, the inventive compounds have therapeutic and cosmetic applications ranging from regulation of neural tissues, bone and cartilage formation and repair, regulation of spermatogenesis, regulation of smooth muscle, regulation of lung, liver and other organs arising from the primative gut, regulation of hematopoietic function, regulation of skin and hair growth, etc. The compounds of the invention can be applied on cells which are provided in culture (in vitro), or on cells in a whole animal (in vivo).
The term "agonist" refers to an agent or analog that binds productively to a receptor and mimics its biological activity. The term "antagonist" refers to an agent that binds to receptors but does not provoke the normal biological response. Thus, an antagonist potentiates or recapitulates, for example, the bioactivity of patched, such as to repress transcription of target genes. The term "Wnt antagonist" as used herein refers not only to any agent that may act by directly inhibiting the normal function of the Wnt protein, but also to any agent that inhibits the Wnt signaling pathway, and thus recapitulates the function of Wnt. The term "Wnt agonist" likewise refers to an agent which antagonizes or blocks the bioactivity of Wnt, such as to increase transcription of target genes.
It is preferred according to the invention to decrease the activity of the Wnt signalling pathway and/or to inhibit the Wnt signalling pathway. Preferably the compounds of the invention are used as Wnt antagonists and used to regulate e.g. proliferation or other biological consequences of mis-expression of Wnt.
As outlined above, an elevated Tcf-beta-catenin level in the nucleus of a cell is a hallmark of an aberrant activation of the Wnt signalling pathway and plays a major role in the development of several kinds of cancer. The measurement of the Tcf-beta-catenin level in the nucleus of the cell may be carried out according procedures known to the person skilled in the art. The measurement of the Tcf-beta-catenin level by means of 6xTcF- lucif erase is described below in the experiments. The use of a compound for modulating the Wnt signalling pathway resulting in a decrease of the relative amount of Tcf-beta- catenin complex in the nucleus of a cell is preferred.
Modulating the Wnt signalling pathway can be carried out by contacting a cell with a compound according to the invention. In one embodiment of the invention, said modulation is performed in vitro or in cell culture. As known to the person skilled in the art, several in vitro and cell culture assays are available.
According to a further embodiment of the invention the modulation can be performed in animals such as mammals. Animal subjects to which the invention is applicable extend to both domestic animals and livestock, raised either as pets or for commercial purposes. Exemplary mammals are mice, rats, guinea pigs, monkeys, dogs and cats. The modulation can also be carried out in humans.
The invention also relates to the compounds (I) to (XVI) of the invention for use as a medicament. The compounds are as defined above; furthermore the embodiments as described below with respect to the use as medicament, e.g. formulation, application and combination, also apply to this aspect of the invention. The pharmaceutical preparation or medicaments comprising the compounds of formulae (I) to (XVI) can be effective for both human and animal subjects. Animal subjects to which the invention is applicable extend to both domestic animals and livestock, raised either as pets or for commercial purposes. Examples are dogs, cats, cattle, horses, sheep, hogs, and goats.
The invention further relates to the use of the compounds of formulae (I) to (XVI) for modulating the Wnt signalling pathway and to the compounds of formulae (I) to (XVI) for modulating the Wnt signalling pathway. The compounds according to the invention are suited for the use for the preparation of a medicament for modulating the Wnt signalling pathway. The present invention provides pharmaceutical preparations or medicaments comprising a compound such as described herein, formulated in an amount sufficient to regulate, in vivo, Wnt pathway, e.g., proliferation or other biological consequences of mis-expression of Wnt.
The invention further relates to the use of a compound according to the invention for the preparation of a medicament for the treatment of a disorder or disease associated with an aberrant activation of Wnt signalling in a mammal and to compounds of the invention for the treatment of a disorder or disease associated with an aberrant activation of Wnt signalling in a mammal. The disorders or diseases associated with the Wnt signalling pathway are for example cell-proliferative disorders, rheumatoid arthritis, diseases connected with aberrant bone density and Dupuytren disease (superficial fibromatosis).
A cell proliferation disorder is a disorder which is connected with some degree of abnormal cell proliferation. Especially, cell-proliferation disorders are important for the development of cancer. A further cell-proliferation disorder is proliferative skin disorders which are marked by unwanted or aberrant proliferation of cutaneous tissue, for example X-linked ichtyosis, psoriasis, atopic dermatitis, allergic contact dermatitis, epidermolytic hyperkeratosis and seborrheic dermatitis. In one preferred aspect, the invention relates to the use of the compounds according to the invention for the preparation of a medicament for the treatment of cancer or proliferative skin disorder.
The Wnt- signalling pathway is also believed to be involved in the maintenance of stem or progenitor cells in a growing list of adult tissues that includes e.g. skin, blood, gut, prostate, muscle and the nervous system. Stem and progenitor cells are important for cell regeneration and consequently for aging and aging related processes. Therefore, the compounds of the invention are useable for the preparation of a medicament for the treatment of aging and age-related disorders and/or diseases.
The compounds of the invention are especially suitable for the use for the preparation of a medicament for the treatment of cancer wherein the cancer is a member of the group multiple myeloma, colon cancer, breast cancer, gastritic cancer, colorectal cancer, lung cancer, prostate cancer, ovarian cancer, bladder cancer, liver cancer, uterine cancer, kidney cancer, leukaemia, gliomas, basal cell carcinoma, rhabdomyosarcoma, mesothelioma, osteosarcoma, medulloblastomas and other primary CNS malignant neuroectodermal tumors.
Possible disorders or diseases which may be treated by administering a medicament prepared from the compounds for formulae (I) to (XVI) are described in detail in US 2007/0219257 Al. Accordingly the compounds of the present invention are applicable to cell culture techniques wherein, whether for genetic or biochemical reasons, the cells have a Wnt receptor. Alternatively, a compound of formulae (I) to (XVI) may be employed in a related method directed towards cells which have a Wnt receptor. In vitro neuronal culture systems have proven to be fundamental and indispensable tools for the study of neural development, as well as the identification of neurotrophic factors such as nerve growth factor (NGF), ciliary trophic factors (CNTF), and brain derived neurotrophic factor (BDNF). One use of the compounds of formulae (I) to (XVI) may be in cultures of neuronal stem cells, such as in the use of such cultures for the generation of new neurons and glia. In such embodiments of the subject method, the cultured cells can be contacted with an aromatic compound of the present invention in order to alter the rate of proliferation of neuronal stem cells in the culture and/or alter the rate of differentiation, or to maintain the integrity of a culture of certain terminally differentiated neuronal cells. In an exemplary embodiment, the subject method can be used to culture, for example, sensory neurons or, alternatively, motorneurons: Such neuronal cultures can be used as convenient assay systems as well as sources of implantable cells for therapeutic treatments.
In another embodiment, the compounds of the invention can be used in the treatment of neoplastic or hyperplastic transformations such as may occur in the central nervous system. For instance, the compounds can be utilized to cause such transformed cells to become either post-mitotic or apoptotic. The compounds may, therefore, be used as part of a treatment for, e.g., malignant gliomas, meningiomas, medulloblastomas, neuroectodermal tumors, and ependymomas. In another embodiment, the compounds of the invention can be used as part of a treatment regimen for malignant meduUoblastoma and other primary CNS malignant neuroectodermal tumors.
In certain embodiments, the compounds of the invention are used as part of treatment program for meduUoblastoma. MeduUoblastoma, a primary brain tumor, is the most common brain tumor in children. A meduUoblastoma is a primitive neuroectodermal tumor (PNET) arising in the posterior fossa. Histologically, they are small round cell tumors commonly arranged in true rosettes, but may display some differentiation to astrocytes, ependymal cells or neurons (Rorke; Kleihues). PNET's may arise in other areas of the brain including the pineal gland (pineoblastoma) and cerebrum.
Medulloblastoma/PNET's are known to recur anywhere in the CNS after resection, and can even metastasize to bone. Pretreatment evaluation should therefore include an examination of the spinal cord to exclude the possibility of "dropped metastases". Gadolinium-enhanced MRI has largely replaced myelography for this purpose, and CSF cytology is obtained postoperatively as a routine procedure. In other embodiments, the compounds of formulae (I) to (XVI) are used as part of a treatment program for hepatocellular carcinoma. Hepatocellular carcinoma is a form of cancer that arises from hepatocytes, the major cell type of the live, and is one of the most common tumors involving mutations in the Wnt pathway. In other embodiments, the compounds of formulae (I) to (XVI) are used as part of treatment program for ependymomas. Ependymomas account for approximately 10% of the pediatric brain tumors in children. Grossly, they are tumors that arise from the ependymal lining of the ventricles and microscopically form rosettes, canals, and perivascular rosettes. Yet another aspect of the present invention concerns the observation in the art that Wnt is involved in morphogenic signals involved in other vertebrate organogenic pathways in addition to neuronal differentiation as described above, having apparent roles in other endodermal patterning, as well as both mesodermal and endodermal differentiation processes. Thus, it is contemplated by the invention that compositions comprising one or more of the inventive compounds can also be utilized for both cell culture and therapeutic uses involving generation and maintenance of non-neuronal tissue. In one embodiment, the present invention makes use of the discovery that Wnt is apparently involved in controlling the development of stem cells responsible for formation of the digestive tract, liver, lungs, and other organs which derive from the primitive gut. Shh serves as an inductive signal from the endoderm to the mesoderm, which is critical to gut morphogenesis. Therefore, for example, compounds of formulae (I) to (XVI) can be employed for regulating the development and maintenance of an artificial liver which can have multiple metabolic functions of a normal liver. In an exemplary embodiment, the compounds of formulae (I) to (XVI) can be used to regulate the proliferation and differentiation of digestive tube stem cells to form hepatocyte cultures which can be used to populate extracellular matrices, or which can be encapsulated in biocompatible polymers, to form both implantable and extracorporeal artificial livers.
In another embodiment, therapeutic compositions of inventive compounds can be utilized in conjunction with transplantation of such artificial livers, as well as embryonic liver structures, to regulate uptake of intraperitoneal implantation, vascularization, and in vivo differentiation and maintenance of the engrafted liver tissue.
In yet another embodiment, the compounds of formulae (I) to (XVI) can be employed therapeutically to regulate such organs after physical, chemical or pathological insult. For instance, therapeutic compositions comprising the compounds of formulae (I) to (XVI) can be utilized in liver repair subsequent to a partial hepatectomy.
The generation of the pancreas and small intestine from the embryonic gut depends on intercellular signalling between the endodermal and mesodermal cells of the gut. In particular, the differentiation of intestinal mesoderm into smooth muscle has been suggested to depend on signals from adjacent endodermal cells. One candidate mediator of endodermally derived signals in the embryonic hindgut is Sonic hedgehog. See, for example, Apelqvist et al., Curr. Biol. 7:801-4 (1997). The Shh gene is expressed throughout the embryonic gut endoderm with the exception of the pancreatic bud endoderm, which instead expresses high levels of the homeodomain protein Ipfl/Pdxl (insulin promoter factor 1/pancreatic and duodenal homeobox 1), an essential regulator of early pancreatic development. Apelqvist et al., supra, have examined whether the differential expression of Shh in the embryonic gut tube controls the differentiation of the surrounding mesoderm into specialised mesoderm derivatives of the small intestine and pancreas. To test this, they used the promoter of the Ipfl/Pdxl gene to selectively express Shh in the developing pancreatic epithelium. In Ipfl/Pdxl-Shh transgenic mice, the pancreatic mesoderm developed into smooth muscle and interstitial cells of Cajal, characteristic of the intestine, rather than into pancreatic mesenchyme and spleen. Also, pancreatic explants exposed to Shh underwent a similar program of intestinal differentiation. These results provide evidence that the differential expression of endodermally derived Shh controls the fate of adjacent mesoderm at different regions of the gut tube.
In the context of the present invention, it is contemplated therefore that the inventive compounds can be used to control or regulate the proliferation and/or differentiation of pancreatic tissue both in vivo and in vitro.
There are a wide variety of pathological cell pro-liferative and differentiative conditions for which the compounds of the present invention may provide therapeutic benefits, with the general strategy being, for example, the correction of aberrant insulin expression, or modulation of differentiation. More generally, however, the present invention relates to the use of the compounds of formulae (I) to (XVI) for inducing and/or maintaining a differentiated state, enhancing survival and/or affecting proliferation of pancreatic cells, by contacting the cells with the subject inhibitors. For instance, it is contemplated by the invention that, in light of the apparent involvement of Wnt in the formation of ordered spatial arrangements of pancreatic tissues, the compounds of the invention could be used as part of a technique to generate and/or maintain such tissue both in vitro and in vivo. For instance, modulation of the function of Wnt can be employed in both cell culture and therapeutic uses involving generation and maintenance β-cells and possibly also for non- pancreatic tissue, such as in controlling the development and maintenance of tissue from the digestive tract, spleen, lungs, colon, and other organs which derive from the primitive gut.
In an exemplary embodiment, the compounds of the invention can be used in the treatment of hyperplastic and neoplastic disorders effecting pancreatic tissue, particularly those characterized by aberrant proliferation of pancreatic cells. For instance, pancreatic cancers are marked by abnormal proliferation of pancreatic cells which can result in alterations of insulin secretory capacity of the pancreas. For instance, certain pancreatic hyperplasias, such as pancreatic carcinomas, can result in hypoinsulinemia due to dysfunction of β-cells or decreased islet cell mass. To the extent that aberrant Wnt signaling may be indicated in disease progression, the compounds of the invention can be used to enhance regeneration of the tissue after anti-tumor therapy.
Moreover, manipulation of Wnt signaling properties at different points may be useful as part of a strategy for reshaping/repairing pancreatic tissue both in vivo and in vitro. In one embodiment, the present invention makes use of the apparent involvement of Wnt in regulating the development of pancreatic tissue. In general, the compounds of the invention can be employed therapeutically to regulate the pancreas after physical, chemical or pathological insult. In yet another embodiment, the subject method can be applied to cell culture techniques, and in particular, may be employed to enhance the initial generation of prosthetic pancreatic tissue devices. Manipulation of proliferation and differentiation of pancreatic tissue, for example, by altering Wnt, can provide a means for more carefully controlling the characteristics of a cultured tissue. Early progenitor cells to the pancreatic islets are multipotential, and apparently coactivate all the islet-specific genes from the time they first appear. As development proceeds, expression of islet- specific hormones, such as insulin, becomes restricted to the pattern of expression characteristic of mature islet cells. The phenotype of mature islet cells, however, is not stable in culture, as reappearance of embryonal traits in mature β-cells can be observed. By utilizing the compounds of the invention, the differentiation path or proliferative index of the cells can be regulated.
Furthermore, manipulation of the differentiative state of pancreatic tissue can be utilized in conjunction with transplantation of artificial pancreas so as to promote implantation, vascularization, and in vivo differentiation and maintenance of the engrafted tissue. For instance, manipulation of Wnt function to affect tissue differentiation can be utilized as a means of maintaining graft viability.
Bellusci et al., Development 124:53 (1997) report that Sonic hedgehog regulates lung mesenchymal cell proliferation in vivo. Accordingly, the compounds of the invention can be used to regulate regeneration of lung tissue, e.g., in the treatment of emphysema.
Fujita et al., Biochem. Biophys. Res. Commun. 238:658 (1997) reported that Sonic hedgehog is expressed in human lung squamous carcinoma and adenocarcinoma cells. The expression of Sonic hedgehog was also detected in the human lung squamous carcinoma tissues, but not in the normal lung tissue of the same patient. They also observed that Sonic hedgehog stimulates the incorporation of BrdU into the carcinoma cells and stimulates their cell growth, while anti-Shh-N inhibited their cell growth. These results suggest that ptc and/or Wnt is involved in the cell growth of such transformed lung tissue and therefore indicates that the compounds of the invention can be used as part of a treatment of lung carcinoma and adenocarcinomas, and other proliferative disorders involving the lung epithelia.
Many other tumors may, based on evidence such as involvement of the Wnt pathway in these tumors, or detected expression of Wnt or its receptors in these tissues during development, be affected by treatment with the compounds of the invention. Such tumors include, but are by no means limited to, tumors related to Gorlin's syndrome (e.g., basal cell carcinoma, meduUoblastoma, meningioma, etc.), tumors evidenced in pet knock-out mice (e.g., hemangioma, rhabdomyosarcoma, etc.), tumors resulting from gli-1 amplification (e.g., glioblastoma, sarcoma, etc.), tumors connected with TRC8, a ptc homolog (e.g., renal carcinoma, thyroid carcinoma, etc.), Ext-l-related tumors (e.g., bone cancer, etc.), Shh-induced tumors (e.g., lung cancer, chon-drosarcomas, etc.), and other tumors (e.g., breast cancer, urogenital cancer (e.g., kidney, bladder, ureter, prostate, etc.), adrenal cancer, gastrointestinal cancer (e.g., stomach, intestine, etc.), etc.).
In still another embodiment of the present invention, compositions comprising one or more of the compounds of the invention can be used in the in vitro generation of skeletal tissue, such as from skeleto genie stem cells, as well as the in vivo treatment of skeletal tissue deficiencies. The present invention particularly contemplates the use of compounds of the invention to regulate the rate of chondrogenesis and/or osteogenesis. By "skeletal tissue deficiency", it is meant a deficiency in bone or other skeletal connective tissue at any site where it is desired to restore the bone or connective tissue, no matter how the deficiency originated, e.g. whether as a result of surgical intervention, removal of tumor, ulceration, implant, fracture, or other traumatic or degenerative conditions.
For instance, the compounds of the invention can be used as part of a regimen for restoring cartilage function to a connective tissue. They are useful in, for example, the repair of defects or lesions in cartilage tissue which is the result of degenerative wear such as that which results in arthritis, as well as other mechanical derangements which may be caused by trauma to the tissue, such as a displacement of torn meniscus tissue, meniscectomy, a laxation of a joint by a torn ligament, malignment of joints, bone fracture, or by hereditary disease. The compounds of the invention may also be useful for remodeling cartilage matrix, such as in plastic or reconstructive surgery, as well as periodontal surgery. They may also be applied to improving a previous reparative procedure, for example, following surgical repair of a meniscus, ligament, or cartilage. Furthermore, it may prevent the onset or exacerbation of degenerative disease if applied early enough after trauma.
One embodiment of the present invention relates to the treating of the afflicted connective tissue with a therapeutically effective amount of compounds of the invention to regulate a cartilage repair response in the connective tissue by managing the rate of differentiation and/or proliferation of chondrocytes embedded in the tissue. Such connective tissues as articular cartilage, interarticular cartilage (menisci), costal cartilage (connecting the true ribs and the sternum), ligaments, and tendons are particularly amenable to treatment in reconstructive and/or regenerative therapies using the subject method. As used herein, regenerative therapies include treatment of degenerative states which have progressed to the point of which impairment of the tissue is obviously manifest, as well as preventive treatments of tissue where degeneration is in its earliest stages or imminent. In an illustrative embodiment, the compounds of the invention can be used as part of a therapeutic intervention in the treatment of cartilage of a diarthroidal joint, such as a knee, an ankle, an elbow, a hip, a wrist, a knuckle of either a finger or toe, or a tempomandibular joint. The treatment can be directed to the meniscus of the joint, to the articular cartilage of the joint, or both. To further illustrate, the compounds of the invention can be used to treat a degenerative disorder of a knee, such as which might be the result of traumatic injury (e.g., a sports injury or excessive wear) or osteoarthritis. The compounds of the invention may be administered as an injection into the joint with, for instance, an arthroscopic needle. In some instances, the injected agent can be in the form of a hydrogel or other slow release vehicle described above in order to permit a more extended and regular contact of the agent with the treated tissue.
The present invention further contemplates the use of the compounds of the invention in the field of cartilage transplantation and prosthetic device therapies. However, problems arise, for instance, because the characteristics of cartilage and fibrocartilage vary between different tissue: such as between articular, meniscal cartilage, ligaments, and tendons, between the two ends of the same ligament or tendon, and between the superficial and deep parts of the tissue. The zonal arrangement of these tissues may reflect a gradual change in mechanical properties, and failure occurs when implanted tissue, which has not differentiated under those conditions, lacks the ability to appropriately respond. For instance, when meniscal cartilage is used to repair anterior cruciate ligaments, the tissue undergoes a metaplasia to pure fibrous tissue. By regulating the rate of chondrogenesis, the compounds of the invention can be used to particularly address this problem, by helping to adaptively control the implanted cells in the new environment and effectively resemble hypertrophic chondrocytes of an earlier developmental stage of the tissue. In similar fashion, the compounds of the invention can be applied to enhancing both the generation of prosthetic cartilage devices and to their implantation. The need for improved treatment has motivated research aimed at creating new cartilage that is based on collagen- glycosaminogly-can templates (Stone et al., Clin. Orthop. Relat. Red 252: 129 (1990)), isolated chondrocytes (Grande et al., J. Orthop. Res. 7:208 (1989); and Takigawa et al., Bone Miner 2:449 (1987)), and chondrocytes attached to natural or synthetic polymers (Walitani et al., J. Bone Jt. Surg. 71B:74 (1989); Vacanti et al., Plast. Reconstr. Surg. 88:753 (1991); von Schroeder et al. J. Biomed. Mater. Res. 25:329 (1991); Freed et al., J. Biomed. Mater. Res. 27: 11 (1993); and the Vacanti et al. U.S. Pat. No.5,041,138). For example, chondrocytes can be grown in culture on biodegradable, biocompatible highly porous scaffolds formed from polymers such as polyglycolic acid, polylactic acid, agarose gel, or other polymers which degrade over time as function of hydrolysis of the polymer backbone into innocuous monomers. The matrices are designed to allow adequate nutrient and gas exchange to the cells until engraftment occurs. The cells can be cultured in vitro until adequate cell volume and density has developed for the cells to be implanted. One advantage of the matrices is that they can be cast or molded into a desired shape on an individual basis, so that the final product closely resembles the patient's own ear or nose (by way of example), or flexible matrices can be used which allow for manipulation at the time of implantation, as in a joint. In one embodiment of the invention, the implants are contacted with a subject aromatic compound during certain stages of the culturing process in order to manage the rate of differentiation of chondrocytes and the formation of hypertrophic chrondrocytes in the culture. In another embodiment, the implanted device is treated with a compound of formulae (I) to (XVI) in order to actively remodel the implanted matrix and to make it more suitable for its intended function. As set out above with respect to tissue transplants, the artificial transplants suffer from the same deficiency of not being derived in a setting which is comparable to the actual mechanical environment in which the matrix is implanted. The ability to regulate the chondrocytes in the matrix by the subject method can allow the implant to acquire characteristics similar to the tissue for which it is intended to replace. In yet another embodiment, the compounds of the invention are used to enhance attachment of prosthetic devices. To illustrate, the compounds of the invention can be used in the implantation of a periodontal prosthesis, wherein the treatment of the surrounding connective tissue stimulates formation of periodontal ligament about the prosthesis.
In other embodiments, the compounds of the invention can be employed as part of a regimen for the generation of bone (osteogenesis) at a site in the animal where such skeletal tissue is deficient. Indian hedgehog (Ihh) is particularly associated with the hypertrophic chondrocytes that are ultimately replaced by osteoblasts. For instance, administration of a compound of the present invention can be employed as part of a method for regulating the rate of bone loss in a subject. For example, preparations comprising subject compounds can be employed, for example, to control endoch-ondral ossification in the formation of a "model" for ossification.
The compounds of the invention also have wide applicability to the treatment or prophylaxis of disorders afflicting epithelial tissue, as well as in cosmetic uses. In general, this includes a step of administering to an animal an amount of a subject aromatic compound effective to alter the growth state of a treated epithelial tissue. The mode of administration and dosage regimens will vary depending on the epithelial tissue(s) which is to be treated. For example, topical formulations will be preferred where the treated tissue is epidermal tissue, such as dermal or mucosal tissues.
Despite significant progress in reconstructive surgical techniques, scarring can be an important obstacle in regaining normal function and appearance of healed skin. This is particularly true when pathologic scarring such as keloids or hypertrophic scars of the hands or face causes functional disability or physical deformity. In the severest circumstances, such scarring may precipitate psychosocial distress and a life of economic deprivation. Wound repair includes the stages of hemostasis, inflammation, proliferation, and remodeling. The proliferative stage involves multiplication of fibroblasts and endothelial and epithelial cells. Through the use of the compounds of the invention, the rate of proliferation of epithelial cells in and proximal to the wound can be controlled in order to accelerate closure of the wound and/or minimize the formation of scar tissue. The present treatment can also be effective as part of a therapeutic regimen for treating oral and paraoral ulcers, e.g., resulting from radiation and/or chemotherapy. Such ulcers commonly develop within days after chemotherapy or radiation therapy. These ulcers usually begin as small, painful irregularly shaped lesions usually covered by a delicate gray necrotic membrane and surrounded by inflammatory tissue. In many instances, a lack of treatment results in proliferation of tissue around the periphery of the lesion on an inflammatory basis. For instance, the epithelium bordering the ulcer usually demonstrates proliferative activity, resulting in loss of continuity of surface epithelium. These lesions, because of their size and loss of epithelial integrity, dispose the body to potential secondary infection. Routine ingestion of food and water becomes a very painful event and, if the ulcers proliferate throughout the alimentary canal, diarrhea usually is evident with all its complicating factors. According to the present invention, a treatment for such ulcers which includes application of a compound of formulae (I) to (XVI) can reduce the abnormal proliferation and differentiation of the affected epithelium, helping to reduce the severity of subsequent inflammatory events. The compounds of the invention and compositions thereof can also be used to treat wounds resulting from dermatological diseases, such as lesions resulting from autoimmune disorders such as psoriasis. Atopic dermititis refers to skin trauma resulting from allergies associated with an immune response caused by allergens such as pollens, foods, dander, insect venoms and plant toxins.
In other embodiments, antiproliferative preparations of subject compounds can be used to inhibit lens epithelial cell proliferation to prevent post-operative complications of extracapsular cataract extraction. Cataract is an intractable eye disease and various studies on a treatment of cataract have been made. But at present, the treatment of cataract is attained by surgical operations. Cataract surgery has been applied for a long time and various operative methods have been examined. Extracapsular lens extraction has become the method of choice for removing cataracts. The major medical advantages of this technique over intra-capsular extraction are lower incidence of aphakic cystoid macular edema and retinal detachment. Extracapsular extraction is also required for implantation of posterior chamber type intraocular lenses which are now considered to be the lenses of choice in most cases.
However, a disadvantage of extracapsular cataract extraction is the high incidence of posterior lens capsule opacification, often called after-cataract, which can occur in up to 50% of cases within three years after surgery. After-cataract is caused by proliferation of equatorial and anterior capsule lens epithelial cells which remain after extracapsular lens extraction. These cells proliferate to cause Sommerling rings, and along with fibroblasts which also deposit and occur on the posterior capsule, cause opacification of the posterior capsule, which interferes with vision. Prevention of after-cataract would be preferable to treatment. To inhibit secondary cataract formation, the present invention provides a means for inhibiting proliferation of the remaining lens epithelial cells. For example, such cells can be induced to remain quiescent by instilling a solution containing a preparation of a compound of formulae (I) to (XVI) into the anterior chamber of the eye after lens removal. Furthermore, the solution can be osmotically balanced to provide minimal effective dosage when instilled into the anterior chamber of the eye, thereby inhibiting subcapsular epithelial growth with some specificity.
The compounds of the invention can also be used in the treatment of corneopathies marked by corneal epithelial cell proliferation, as for example in ocular epithelial disorders such as epithelial downgrowth or squamous cell carcinomas of the ocular surface.
Yet another aspect of the present invention relates to the use of the compounds of the invention to control hair growth. Hair is basically composed of keratin, a tough and insoluble protein; its chief strength lies in its disulphide bond of cystine. Each individual hair comprises a cylindrical shaft and a root, and is contained in a follicle, a flask-like depression in the skin. The bottom of the follicle contains a finger-like projection termed the papilla, which consists of connective tissue from which hair grows, and through which blood vessels supply the cells with nourishment. The shaft is the part that extends outwards from the skin surface, whilst the root has been described as the buried part of the hair. The base of the root expands into the hair bulb, which rests upon the papilla. Cells from which the hair is produced grow in the bulb of the follicle; they are extruded in the form of fibers as the cells proliferate in the follicle. Hair "growth" refers to the formation and elongation of the hair fiber by the dividing cells.
As is well known in the art, the common hair cycle is divided into three stages: anagen, catagen and telogen. During the active phase (anagen), the epidermal stem cells of the dermal papilla divide rapidly. Daughter cells move upward and differentiate to form the concentric layers of the hair itself. The transitional stage, catagen, is marked by the cessation of mitosis of the stem cells in the follicle. The resting stage is known as telogen, where the hair is retained within the scalp for several weeks before an emerging new hair developing below it dislodges the telogen-phase shaft from its follicle. From this model it has become clear that the larger the pool of dividing stem cells that differentiate into hair cells, the more hair growth occurs. Accordingly, methods for increasing or reducing hair growth can be carried out by potentiating or inhibiting, respectively, the proliferation of these stem cells. In certain embodiments, the compounds of the invention can be employed as a way of reducing the growth of human hair as opposed to its conventional removal by cutting, shaving, or depilation. For instance, the present method can be used in the treatment of trichosis characterized by abnormally rapid or dense growth of hair, e.g. hypertrichosis. In an exemplary embodiment, subject compounds can be used to manage hirsutism, a disorder marked by abnormal hairiness. The compounds of the invention can also provide a process for extending the duration of depilation.
Moreover, because a subject compound will often be cytostatic to epithelial; cells, rather than cytotoxic, such agents can be used to protect hair follicle cells from cytotoxic agents which require progression into S-phase of the cell-cycle for efficacy, e.g. radiation-induced death. Treatment by the compounds of the invention can provide protection by causing the hair follicle cells to become quiescent, e.g., by inhibiting the cells from entering S phase, and thereby preventing the follicle cells from undergoing mitotic catastrophe or programmed cell death. For instance, compounds of the invention can be used for patients undergoing chemo- or radiation-therapies which ordinarily result in hair loss. By inhibiting cell-cycle progression during such therapies, the subject treatment can protect hair follicle cells from death which might otherwise result from activation of cell death programs. After the therapy has concluded, the administration of compounds of the invention can be stopped with concommitant relief of the inhibition of follicle cell proliferation.
The compounds of the invention can also be used in the treatment of folliculitis, such as folliculitis decalvans, folliculitis ulerythematosa reticulata or keloid folliculitis. For example, a cosmetic preparation of a compound of formulae (I) to (XVI) can be applied topically in the treatment of pseudofolliculitis, a chronic disorder occurring most often in the submandibular region of the neck and associated with shaving, the characteristic lesions of which are erythematous papules and pustules containing buried hairs.
In another aspect of the invention, the compounds of the invention can be used to induce differentiation and/or inhibit proliferation of epithelially derived tissue. Such forms of these molecules can provide a basis for differentiation therapy for the treatment of hyperplastic and/or neoplastic conditions involving epithelial tissue. For example, such preparations can be used for the treatment of cutaneous diseases in which there is abnormal proliferation or growth of cells of the skin.
For instance, the pharmaceutical preparations of the invention are intended for the treatment of hyperplastic epidermal conditions, such as keratosis, as well as for the treatment of neoplastic epidermal conditions such as those characterized by a high proliferation rate for various skin cancers, as for example basal cell carcinoma or squamous cell carcinoma. The compounds of the invention can also be used in the treatment of autoimmune diseases affecting the skin, in particular, of dermatological diseases involving morbid proliferation and/or keratinization of the epidermis, as for example, caused by psoriasis or atopic dermatosis.
Many common diseases of the skin, such as psoriasis, squamous cell carcinoma, keratoacanthoma and actinic keratosis are characterized by localized abnormal proliferation and growth. For example, in psoriasis, which is characterized by scaly, red, elevated plaques on the skin, the keratinocytes are known to proliferate much more rapidly than normal and to differentiate less completely.
In one embodiment, the preparations of the present invention are suitable for the treatment of dermatological ailments linked to keratinization disorders causing abnormal proliferation of skin cells, which disorders may be marked by either inflammatory or noninflammatory components. To illustrate, therapeutic preparations of an inventive compound, e.g., which promotes quiescense or differentiation, can be used to treat varying forms of psoriasis, be they cutaneous, mucosal orungual. Psoriasis, as described above, is typically characterized by epidermal keratinocytes which display marked proliferative activation and differentiation along a "regenerative" pathway. Treatment with an antiproliferative compound of the invention can be used to reverse the pathological epidermal activiation and can provide a basis for sustained remission of the disease. A variety of other keratotic lesions are also candidates for treatment with the compounds of the invention. Actinic keratoses, for example, are superficial inflammatory premalig-nant tumors arising on sun-exposed and irradiated skin. The lesions are erythematous to brown with variable scaling. Current therapies include excisional and cryosurgery. These treatments are painful, however, and often produce cosmetically unacceptable scarring. Accordingly, treatment of keratosis, such as actinic keratosis, can include application, preferably topical, of a composition containing at least one compound of formulae (I) to (XVI) in amounts sufficient to inhibit hyperproliferation of epidermal/epidermoid cells of the lesion.
Acne represents yet another dermatologic ailment which may be treated by the compounds of the invention. Acne vulgaris, for instance, is a multifactorial disease most commonly occurring in teenagers and young adults, and is characterized by the appearance of inflammatory and noninflammatory lesions on the face and upper trunk. The basic defect which gives rise to acne vulgaris is hypercornification of the duct of a hyperactive sebaceous gland. Hypercornification blocks the normal mobility of skin and follicle microorganisms, and in so doing, stimulates the release of lipases by Propinobac-terium acnes and Staphylococcus epidermidis bacteria and Pitrosporum ovale, a yeast. Treatment with an antiproliferative compound of formulae (I) to (XVI) , particularly topical preparations, may be useful for preventing the transitional features of the ducts, e.g. hypercornification, which lead to lesion formation. The subject treatment may further include, for example, antibiotics, retinoids and antiandrogens.
The present invention also provides a method for treating various forms of dermatitis. Dermatitis is a descriptive term referring to poorly demarcated lesions which are either pruritic, erythematous, scaly, blistered, weeping, fissured or crusted. These lesions arise from any of a wide variety of causes. The most common types of dermatitis are atopic, contact and diaper dermatitis. For instance, seborrheic dermatitis is a chronic, usually pruritic, dermatitis with erythema, dry, moist, or greasy scaling, and yellow crusted patches on various areas, especially the scalp, with exfoliation of an excessive amount of dry scales. The compounds of the invention can also be used in the treatment of stasis dermatitis, an often chronic, usually eczematous dermatitis. Actinic dermatitis is dermatitis that due to exposure to actinic radiation such as that from the sun, ultraviolet waves or x-or gamma-radiation. According to the present invention, the compounds of the invention can be used in the treatment and/or prevention of certain symptoms of dermatitis caused by unwanted proliferation of epithelial cells. Such therapies for these various forms of dermatitis can also include topical and systemic corticosteroids, antipuritics, and antibiotics.
Ailments which may be treated by the compounds of the invention are disorders specific to non-humans, such as mange. In still another embodiment, the compounds of the invention can be used in the treatment of human cancers, particularly basal cell carcinomas and other tumors of epithelial tissues such as the skin. For example, compounds of the invention can be employed, in the subject method, as part of a treatment for basal cell nevus syndrome (BCNS), and other other human carcinomas, adenocarcinomas, sarcomas and the like.
In another embodiment, the compounds of the invention are used as part of a treatment of prophylaxis regimen for treating (or preventing) basal cell carcinoma. The deregulation of the Wnt signaling pathway may be a general feature of basal cell carcinomas caused by ptc mutations. Consistent overexpres-sion of human ptc mRNA has been described in tumors of familial and sporadic BCCs, determined by in situ hybridization. Mutations that inactivate ptc may be expected to result in overexpression of mutant Ptc, because ptc displays negative autoregulation. Likewise, mutations that inactivate Wnt may be expected to result in overexpression of mutant Wnt, because Wnt displays negative autoregulation. Prior research demonstrates that overexpression of hedgehog proteins can also lead to tumorigenesis. That sonic hedgehog (Shh) has a role in tumorigenesis in the mouse has been suggested by research in which transgenic mice overexpressing Shh in the skin developed features of BCNS, including multiple BCC-like epidermal proliferations over the entire skin surface, after only a few days of skin development. A mutation in the Shh human gene from a BCC was also described; it was suggested that Wnt, Shh or other Hh genes in humans could act as dominant oncogenes in humans. Sporadic ptc mutations have also been observed in BCCs from otherwise normal individuals, some of which are UV- signature mutations. In one recent study of sporadic BCCs, five UV- signature type mutations, either CT or CCTT changes, were found out of fifteen tumors determined to contain ptc mutations. Another recent analysis of sporadic ptc mutations in BCCs and neuroectodermal tumors revealed one CT change in one of three ptc mutations found in the BCCs. See, for example, Goodrich et al., Science 277: 1109-13 (1997); Xie et al. Cancer Res. 57:2369-72 (1997); Oro et al. Science 276:817-21 (1997); Xie et al, Genes Chromosomes Cancer 18:305-9 (1997); Stone et al, Nature 384: 129-34 (1996); and Johnson et al. Science 272: 1668-71 (1996).
The compounds of the invention can also be used to treat patients with BCNS, e.g., to prevent BCC or other effects of the disease which may be the result of Wnt-mediated disorders. Basal cell nevus syndrome is a rare autosomal dominant disorder characterized by multiple BCCs that appear at a young age. BCNS patients are very susceptible to the development of these tumors; in the second decade of life, large numbers appear, mainly on sun-exposed areas of the skin. This disease also causes a number of developmental abnormalities, including rib, head and face alterations, and sometimes Polydactyly, syndactyly, and spina bifida. They also develop a number of tumor types in addition to BCCs: fibromas of the ovaries and heart, cysts of the skin and jaws, and in the central nervous system, medulloblas-tomas and meningiomas. The compounds of the invention can be used to prevent or treat such tumor types in BCNS and non-BCNS patients. Studies of BCNS patients show that they have both genomic and sporadic mutations in the ptc gene, suggesting that these mutations are the ultimate cause of this disease. In another aspect, the present invention provides pharmaceutical preparations and methods for controlling the formation of megakaryocyte-derived cells and/or controlling the functional performance of megakaryocyte-derived cells. For instance, certain of the compositions disclosed herein may be applied to the treatment or prevention of a variety hyperplastic or neoplastic conditions affecting platelets.
Furthermore, the invention relates to pharmaceutical compositions comprising at least one compound according to the invention. In a preferred embodiment, the invention relates to pharmaceutical compositions comprising at least one compound according to the invention in a mixture with an inert carrier, where said inert carrier is a pharmaceutical carrier.
The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the compound is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, including but not limited to peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered orally. Saline and aqueous dextrose are preferred carriers when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions are preferably employed as liquid carriers for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, sustained-release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin. Such compositions will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.
The invention further relates to a process for the preparation of a medicament comprising the steps of: a) preparing at least one compound according to the formulae (I) to (XVI); and b) formulating a medicament containing at least said compound; and to a method of treating a mammal for modulating the Wnt signalling pathway wherein the method comprises administering to said mammal a therapeutically effective amount of a compound according to formulae (I) to (XVI).
The compounds according to the invention used for the preparation of a medicament for the modulation of the Wnt signalling pathway in a mammal may be administered in any convenient route. The compounds are formulated to be compatible with the desired route of administration and may be administered together with other biologically active agents.
The compounds may be formulated for the intravenous, intradermal, subcutanus, intramuscular, intraperitoneal, epidural, oral, transdermal, transmucosal, rectal or pulmonary administration. Administration can be systemic or local. Pulmonary administration can be employed by use of an inhaler or nebulizer, and formulation with an aerosolizing agent, for example. In another embodiment, the compound can be delivered in a vesicle, in particular a liposome (Langer (1990) Science 249, 1527. In yet another embodiment, the compound can be delivered via a controlled release system. In one embodiment, a pump may be used (Sefton (1987) CRC Crit. Ref. Biomed. Eng. 14, 201; Buchwald et al. (1980) Surgery 88, 507; Saudek et al. (1989) N. Engl. J. Med. 321, 574). In another embodiment, polymeric materials can be used (Ranger and Peppas (1983) Macromol. Sci. Rev. Macromol. Chem. 23, 61; Levy et al. (1985) Science 228, 190; During et al. (1989) Ann. Neurol. 25, 351; Howard et al. (1989) J. Neurosurg. 71, 858). In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target, i.e., the brain, thus requiring only a fraction of the systemic dose (e.g. Goodson, 1984, In: Medical Applications of Controlled Release, supra, Vol. 2, 115). Other controlled release systems are discussed in the review by Langer (1990, Science 249, 1527).
Toxicity and therapeutic efficacy of the compounds of the invention can be determined by standard pharmaceutical procedures in cell cultures or experimental animals. The results obtained from the cell culture assays and animal studies can be used in formulating the range of dosage for use in medicaments in humans. The specific dosage for any particular subject is influenced by several factors, e.g. by the activity of the specific compound used, the age, body weight, general health, gender and diet of the subject, the time and the route of administration and the rate of excretion.
Examples
Example 1 6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-pyrazolo[5,l- b]quinazoline-3-carboxylic acid
Figure imgf000042_0001
(1 )
3-Aminopyarzole-4-carboxylic acid (5.00 g, 39.3 mmol), tolualdehyde (5.64 g, 47 mmol) and dimedone (6.60 g, 47 mmol) were taken in 2-propanol (50 ml) in a 250 ml RB flask equipped with a magnetic stirrer and refluxed with stirring under LC-MS control for 4 h whereby the reaction was complete.
The reaction mixture was cooled down to room temperature, diluted with cyclohexane (100 ml), stirred for 1 h at room temperature, solids were collected by filtration, washed first with 1: 1 mixture of cyclohexane-diethylether (50 ml) and then with ice cold water and dried under high vacuum to yield 10.8 g (30.7 mmol, 78.1%) of the pure white solids of the title compound.
Example 2 6,6-Dimethyl-8-oxo-9-p-tolyl-5,6,7,8-tetrahydro-pyrazolo[5,l- ]quinazoline-3-carboxylic acid
Figure imgf000043_0001
6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-pyrazolo[5,l-b]quinazoline-3- carboxylic acid (14.2 g, 40.1 mmol, see example 1) were suspended in acetone (300 ml) in a 500 ml Round bottomed flask with a magnetic stirrer and then CAN (48.4 g, 88.2 mmol) dissolved in water (80 ml) was added drop wise at room temperature under vigorous stirring. The mixture was stirred at room temperature for lh under LC-MS control whereby the entire compound went to solution giving an orange solution.
The reaction mixture was partitioned between dichloromethane (300 ml) and water (300ml), organic phase was separated, dried over magnesium sulfate, solvent was removed under vacuum and crystallized from ether to get yellow solids of the title compound (13.0 g, 37.2 mmol, 92.9%). Example 3 6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-pyrazolo[5,l- b]quinazoline-3-carboxylic acid ethyl ester
Figure imgf000044_0001
(3)
3-Aminopyarzole-4-carboxylic acid ethyl ester (31.2 g, 200 mmol), tolualdehyde (26 ml, 220 mmol), dimedone (31 g, 220 mol) and acetic acid (5 ml) were taken in toluene (250 ml) in a 500 ml RB flask equipped with a magnetic stirrer and refluxed with stirring under LC-MS control for 8h whereby the reaction was complete.
The reaction mixture was cooled down to room temperature, diluted with cyclohexane (250 ml), stirred for 2 h, solids were collected by filtration, washed first with 1: 1 mixture of cyclohexane-diethylether (50 ml) and then with 40% aqueous 2-propanol (100 ml) and dried under high to yield 66.4 g (175 mmol, 87.6%) of the pure white solids of the title compound.
Figure imgf000044_0002
(3) (4) 6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-pyrazolo[5,l-b]quinazoline-3- carboxylic acid ethyl ester (9.5 g, 25 mmol, see example 3) was suspended in acetonitrile (100 ml) in a 250ml RB flask equipped with a magnetic stirrer, charged with potassium carbonate (3.45g, 25 mmol) and methyl iodide (7.0g, 50 mmol) and then stirred at 70 °C for 3 h under LC-MS control.
The reaction mixture was cooled down to room temperature, excess potassium carbonate was neutralized with acetic, partitioned between DCM (100 ml) and water (100 ml), organic phase was separated, dried over magnesium sulfate, solvent was removed under vacuum a 40 °C and then dried under high vacuum to yield 9.6 g (24.4 mmol, 97.7%) of the title compound as pale yellow solid.
Example 5 4,6,6-Trimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-pyrazolo[5,l- b]quinazoline-3-carboxylic acid
Figure imgf000045_0001
4,6,6-Trimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-pyrazolo[5,l-b]quinazoline-3- carboxylic acid ethyl ester (9.5g, 24.1 mmol, see example 4) was taken in 100 ml methanol in a 250 ml RB flask equipped with a magnetic stirrer at room temperature and then charged with 2M aq. sodium hydroxide (100 ml) under vigorous stirring and stirred further at room temperature for 4h under LC-MS control.
The reaction mixture was neutralized with concentrated hydrochloric acid, methanol was removed under vacuum at 40 °C, stored at 4 °C for 2h, solids collected by filtration, washed with ice cold water and dried under high vacuum to yield pure white solids (8.6g, 23.56 mmol, 97.8%) of the title compound. 4-AUyl-6,6-dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-pyrazolo[5,l- b]quinazoline-3-carboxylic acid ethyl ester
Figure imgf000046_0001
(4) (6)
6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-pyrazolo[5,l-b]quinazoline-3- carboxylic acid ethyl ester (9.5 g, 25 mmol, see example 3) was suspended in acetonitrile (100 ml) in a 250ml RB flask equipped with a magnetic stirrer, charged with potassium carbonate (3.45g, 25 mmol) and allyl bromide (3.63g, 30 mmol) and then stirred at 70 °C for 5 h under LC-MS control.
The reaction mixture was cooled down to room temperature, excess potassium carbonate was neutralized with acetic, partitioned between DCM (100 ml) and water (100 ml), organic phase was separated, dried over magnesium sulfate, solvent was removed under vacuum a 40 °C and then dried under high vacuum to yield 10.3 g (24.5 mmol, 98%) of the title compound as white solid.
Example 7 4-AUyl-6,6-dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-pyrazolo[5,l- b]quinazoline-3-carboxylic acid
Figure imgf000047_0001
4-Allyl-6,6-dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-pyrazolo[5,l-b]quinazoline-3- carboxylic acid ethyl ester (lOg, 23.83 mmol, see example 6) was taken in 100 ml methanol in a 250 ml RB flask equipped with a magnetic stirrer at room temperature and then charged with 2M aq. sodium hydroxide (100 ml) under vigorous stirring and stirred further at room temperature for 4h by monitoring the reaction with LC-MS.
It was then neutralized with concentrated hydrochloric acid, methanol was removed under vacuum at 40 °C, stored at 4°C for 2h, solids collected by filtration, washed with ice cold water and dried under vacuum to yield pure yellowish solids (8.3g, 21.2 mmol, 89.2%) of the title compound.
Example 8 6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-pyrazolo[5,l- b]quinazoline-3-carbonitrile
Figure imgf000048_0001
5,5-Dimethyl-l,3cyclohexanedione (31g, 221mmol), tolualdehyde (26ml, 220 mmol) and 5-Amino-lH-pyrazole-4-carbonitrile 21.6g, 200 mmol) were taken in 250 ml dry toluene with 5 ml AcOH in 500 ml RB flask equipped with a magnetic stirrer and refluxed for 4h with stirring under LC-MS monitoring of the reaction.
Cooled down to room temperature, diluted with cyclohexane (250 ml), stirred for lh at room temperature, solids collected by filtration, washed with ether/c-hexane (100ml, 1: 1 mixture), washed with 40% aq. 2-PrOH (100ml) and then dried to get pure white solids (49g, 147.3 mmol, 73.7%) of the product.
Example 9 6,6-Dimethyl-8-oxo-9-p-tolyl-5,6,7,8-tetrahydro-pyrazolo[5,l- b]quinazoline-3-carbonitrile
Figure imgf000049_0001
(8) (9)
6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-pyrazolo[5,l-b]quinazoline-3- carbonitrile (18.3 g, 55.1 mmol, see example 8) were suspended in acetone (300 ml) in a RB flask equipped with a magnetic stirrer and then CAN (66.5 g, 121.2 mmol) dissolved in water (80 ml) was added drop wise at room temperature under vigorous stirring. The mixture was stirred at room temperature for lh under LC-MS control.
The reaction mixture was diluted with ice cold water (500 ml), solids were collected by filtration and washed with water to get light yellow crystals of the title compound (15.8 g, 47.8 mmol, 86.8%).
Example 10 (3- Cyano-6,6-dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-tetrahydro-6H- pyrazolo[5,l-b]quinazolin-4-yl)-acetic acid ethyl ester
Figure imgf000050_0001
6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-pyrazolo[5,l-b]quinazoline-3- carbonitrile (10 g, 30 mmol, see example 8), triethylamine (8.28 ml, 60 mmol) and ethyl chlroacetate (6.36 ml, 60 mmol) were taken in dry methylnitrile (100 ml) in a 250 ml RB flask equipped with a magnetic stirrer and stirred at 70 °C for 18 h and the reaction was monitored by LC-MS.
The reaction mixture was cooled down to room temperature, excess potassium carbonate was neutralized with acetic, partitioned between DCM (100 ml) and water (100 ml), organic phase was separated, dried over magnesium sulfate, solvent was removed under vacuum a 40 °C and then dried under high vacuum to yield 11.5 g (27.5 mmol, 91.6%) of the title compound as white solid.
Example 11 (3- Cyano-6,6-dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-tetrahydi
pyrazolo[5,l-b]quinazolin-4-yl)-acetic acid
Figure imgf000051_0001
(3-Cyano-6,6-dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-tetrahydro-6H-pyrazolo[5,l- b]quinazolin-4-yl)-acetic acid ethyl ester (11.5g, 27.5 mmol, see example 10) was suspended in 70 ml methanol at room temperature in a 250 ml RB flask and then charged with 2M aq. sodium hydroxide(50 ml) under vigorous stirring and stirred further at the same temperature for 1 h with LC-MS monitoring of the reaction.
It was then neutralized with 10M aq. hydrochloric acid, methanol was removed under vacuum at 40 °C, stored at 4 °C for 2 h, solids were collected by filtration, washed with ice cold water and then dried under high vacuum to yield light yellowish solid (10.5 g, 26.9 mmol, 97.8%) of the title compound.
Example 12 6,6-Dimethyl-9-p-tolyl-4,5,6,7,8,9-tetrahydro-4H-[l,2,4]triazolo[5,l- b]quinazolin-8-one
Figure imgf000052_0001
5,5-Dimethyl-l,3cyclohexanedione (16.8g, 120mmol), tolualdehyde (14ml, 120 mmol) and 3-amino-lH-triazole (8.4g, 100 mmol) were taken in 100 ml dry toluene with 5 ml AcOH in a 250 ml RB flask equipped with a magnetic stirrer and refluxed for 4h with stirring under LC-MS monitoring of the reaction.
It was cooled down to room temperature, diluted with cyclohexane (250 ml), stirred for lh at room temperature, solids collected by filtration, washed with ether/c-hexane (100ml, 1: 1 mixture), washed with 40% aq. 2-PrOH (100ml) and then dried to get pure white solids (28g, 91 mmol, 91%) of the product.
Example 13 (6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-tetrahydro-6H- l,2,4]triazolo[5,l-b]quinazolin-4-yl)-acetic acid ethyl ester
Figure imgf000053_0001
6,6-Dimethyl-9-p-tolyl-4,5,6,7,8,9-tetrahydro-4H-[l,2,4]triazolo[5,l-b]quinazolin-8-one (12.4g, 40 mmol, see example 12) was suspended in 100 ml 1: 1 mixture of DMF/toluene a 250 ml B flask equipped with a magnetic stirrer and then charged with TEA (80 mmol) and ethyl chloroacetate (80 mmol) and stirred at 90 C for 4h.
Toluene was removed under vacuum, diluted with water to 150 ml, stirred at room temperature for lh, solids were collected by filtration, washed with water to get white solids (13.5g, 34.2 mmol, 85.5%) of the title compound.
Example 14 (6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-tetrahydro-6H-l,2,4]triazolo[5,l- b]quinazolin-4-yl)-acetic acid
Figure imgf000053_0002
(6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-tetrahydro-6H-[l,2,4]triazolo[5,l-b]quinazolin- 4-yl)-acetic acid ethyl ester (12g, 30.4 mmol, see example 13) was suspended in 100 ml methanol in a 250 ml RB flask equipped with a magnetic stirrer at room temperature and then charged with 2M aq. sodium hydroxide(100 ml) under vigorous stirring and stirred further at the same temperature for 4 h with LC-MS monitoring of the reaction.
It was then neutralized with 10M aq. hydrochloric acid, methanol was removed under vacuum at 40 °C, stored at 4 °C for 2 h, solids were collected by filtration, washed with ice cold water and then dried under high vacuum to yield light yellowish solid (11 g, 30.0 mmol, 98.7%) of the title compound.
Example 15 6,6-Dimethyl-8-oxo-9-p-tolyl-(4,5, 6,7,8, 9-hexahydro-[l, 2,4]triazolo[5,l- b]quinazoline-2-carboxylic acid methyl ester
Figure imgf000054_0001
5,5-Dimethyl-l,3-cyclohexanedione (14.2g, 100 mmol), tolualdehyde (14ml, 120 mmol) and 5-amino-lH-triazole-3-carboxylic acid methyl ester (16.8g, 120 mmol) were taken in 125 ml dry toluene with 3 ml AcOH in a 250 ml RB flask equipped with a magnetic stirrer and refluxed with stirring for 4h under LC-MS monitoring of the reaction.
It was cooled down to room temperature, diluted with cyclohexane (250 ml), stirred for lh at room temperature, solids collected by filtration, washed with ether/c-hexane (100ml, 1: 1 mixture), washed with 40% aq. 2-PrOH (100ml) and then dried to get pure white solids (23g, 62.8 mmol, 62.8%) of the product. Example 16 6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-[l,2,4]triazolo[5,l- b]quinazoline-2-carboxylic acid
Figure imgf000055_0001
(15) (16)
6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-[l,2,4]triazolo[5,l-b]quinazoline-2- carboxylic acid methyl ester (7.3g, 20 mmol, see example 15) was suspended in 100 ml methanol in a 250 ml RB flask at room temperature and then charged with 2M aq. sodium hydroxide(100 ml) under vigorous stirring and stirred further at the same temperature for 0.5 h with LC-MS monitoring of the reaction.
It was then neutralized with 10M aq. hydrochloric acid, methanol was removed under vacuum at 40 °C, stored at 4 °C for 2 h, solids were collected by filtration, washed with ice cold water and then dried under high vacuum to yield light yellowish solid (7.1 g, 20 mmol, 100%) of the title compound.
Example 17 6,6-Dimethyl-8-oxo-9-p-tolyl-5,6,7,8-tetrahydro-[l,2,4]triazolo[5,l- b]quinazoline-2-carboxylic acid
Figure imgf000055_0002
(16) (17)
6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-[l,2,4]triazolo[5,l-b]quinazoline-2- carboxylic acid (10.2g, 28.94 mmol, see example 16) was taken in acetone in a 250 ml round bottom flask at room temperature and then charged with ammonium cerium(IV) nitrate (CAN, 34.9g, 63.7 mmol) dissolved in 30 ml water by drop wise addition under vigorous stirring. It was then stirred at room temperature under LC-MS control for 3h. The reaction mixture was diluted with water to 250 ml, stored at -20C for 2h, solids were collected by filtration, washed with ice cold water and then dried to yield 7.4g (21.2 mmol, 73.3%) of the pure light yellow solids of the title compound.
Example 18 4,6,6-Trimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-
[l,2,4]triazolo[5,l-b]quinazoline-2-carboxylic acid methyl ester
Figure imgf000056_0001
(15) (18)
6,6-Dimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-[l,2,4]triazolo[5,l-b]quinazoline-2- carboxylic acid methyl ester (12.4g, 33.8 mmol, see example 15), potassium carbonate (3.5g, 25 mmol) and methyl iodide (3.2 ml, 50.7 mmol) were taken in 100 ml dry DMF and stirred at 70 C for 2.5 h.
Excess potassium carbonate was neutralized with acetic acid, diluted with water 400 ml, stirred vigorously for lh, solids were collected by filtration, washed with water and dried under high vacuum to yield light yellowish solids (10.67 g, 29.12 mmol, 86.15%).
Example 19 4,6,6-Trimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro- [1 ,2,4] triazolo [5,1 -b] quinazoline-2-carboxylic acid
Figure imgf000057_0001
(18) (19)
4,6,6-Trimethyl-8-oxo-9-p-tolyl-4,5,6,7,8,9-hexahydro-[l,2,4]triazolo[5,l-b]quinazoline-2- carboxylic acid methyl ester (10.67g, 29.12 mmol, see example 18) was suspended in 50 ml methanol in a 250 ml RB flask equipped with a magnetic stirrer at room temperature and then charged with 2M aq. sodium hydroxide(50 ml) under vigorous stirring and stirred further at the same temperature for 2.5 h with LC-MS monitoring of the reaction.
It was then neutralized with 10M aq. hydrochloric acid, methanol was removed under vacuum at 40 °C, stored at 4 °C for 2 h, solids were collected by filtration, washed with ice cold water and then dried under high vacuum to yield light yellowish solid (10.3 g, 28.2 mmol, 96.5%) of the title compound.
Example 20 7,7-Dimethyl-4-p-tolyl-4,7,8,9-tetrahydro-6H-l,2,3,3a,9-pentaaza- cyclopenta[b]naphthalen-5-one
Figure imgf000057_0002
(20) 5,5-Dimethyl-l,3cyclohexanedione (8.4g, 60mmol), tolualdehyde (7ml, 60 mmol) and aminotetrazole (4.25g, 50 mmol) were taken in 150 ml methanol and refluxed (120 C oil bath temp.) for 2h. Toluene 50 ml was added and stirred further 2h, whereby methanol was completely removed. LC-MS: reaction complete with the desired product as major product. On the completion of the reaction, about 40 ml of the toluene was left. It was then diluted with 2-propanol to about 100 ml and then with diethylether to 200 ml. Stirred for 3h, solids separated by filtration, washed with 2-propanol and dried under high vacuum to yield 7.2g (23.3 mmol, 38.8%) white solids of the title compound. Example 21 (7,7-Dimethyl-5-oxo-4-p-tolyl-5,6,7,8-tetrahydro-4H-l,2,3,3a,9- pentaaza-cyclopenta[b]naphthalen-9-yl)-acetic acid ethyl ester
Figure imgf000058_0001
(20) (21)
7,7-Dimethyl-4-p-tolyl-4,7,8,9-tetrahydro-6H- 1,2,3, 3a,9-pentaaza-yclopenta[b]naphthalen- 5-one (20g, 64.6mmol, see example 20) and ethyl chloroacetate (96.9 mmol) and TEA (96.9 mmol) were taken in acetonitrile (100 ml) in a 250 ml RB flask and stirred at 60 C for 8h under LC-MS control.
The reaction mixture was cooled down to room temperature. It was then partitioned between DCM (200 ml) and water (200 ml), the organic phase was separated, dried over magnesium sulfate and the solvent was evaporated under vacuum at 40 °C to dryness giving 22g (55.6 mmol, 83.7%) of the light yellowish solids of the title product.
Figure imgf000059_0001
(7,7-Dimethyl-5-oxo-4-p-tolyl-5,6,7,8-tetrahydro-4H-l,2,3,3a,9-pentaaza- cyclopenta[b]naphthalen-9-yl)-acetic acid ethyl ester (22g, 23.83 mmol, see example 21) was taken in 100 ml methanol in a RB flask at room temperature and then charged with 2M aq. sodium hydroxide (100 ml) under vigorous stirring and stirred further at the same temperature for 4h under LC-MS control.
It was then neutralized with 10M hydrochloric acid, methanol was removed under vacuum at 40 °C, stored at 4 °C for 2h, solids collected by filtration, washed with water and dried to yield white solids (20g, 54.4 mmol, 96.5%) of the title compound.
Example 23 Benzoic acid N'-[9-(3-bromo-4,5-dimethoxy-phenyl)-6,6-dimethyl-8-oxo-4 ,5,6,7,8,9-hexahydro-pyrazolo[5,l-b]quinazoline-3-carbonyl]-hydrazide
Figure imgf000059_0002
9-(3-Bromo-4,5-dimethoxy-phenyl)-6,6-dimethyl-8-oxo-4,5,6,7,8,9-hexahydro- pyrazolo[5,l-b]quinazoline-3-carboxylic acid 4-nitro-phenyl ester (59.7 mg, 0.1 mmol) was taken in dry DMF (1 ml) under argon and charged with benzoic acid hydrazide (40.8 mg, 0.3 mmol) and stirred at 60 C till the reation was complete.The reaction mixture was diluted with 1 : 1 mixture of DMSO/CAN to 2 ml, filter through a microfilter and the product was purified by HPLC to yield 38 mg of the pure product.
Example 24 4-Methoxy-benzoic acid N'-[9-(3-bromo-4,5-dimethoxy-phenyl)-6,6-imeth yl-8-oxo-4,5,6,7,8,9-hexahydro-pyrazolo[5,l-b]quinazoline-3-carbonyl]-hydrazide
Figure imgf000060_0001
9-(3-Bromo-4,5-dimethoxy-phenyl)-6,6-dimethyl-8-oxo-4,5,6,7,8,9-hexahydro- pyrazolo[5,l-b]quinazoline-3-carboxylic acid 4-nitro-phenyl ester (59.7 mg, 0.1 mmol) was taken in dry DMF (1 ml) under argon and charged with 4-methoxybenzoic acid hydrazide (49.8 mg, 0.3 mmol) and stirred at 60 C till the reation was complete.The reaction mixture was diluted with 1: 1 mixture of DMSO/CAN to 2 ml, filter through a microfilter and the product was purified by HPLC to yield 31 mg of the pure product.
Example 25 Thiophene-2-carboxylic acid N'-[9-(3-bromo-4,5-dimethoxy-phenyl) dimethyl-8-oxo-4,5,6,7,8,9-hexahydro-pyrazolo[5,l-b]quinazoline-3-carbonyl]- hydrazide
Figure imgf000061_0001
9-(3-Bromo-4,5-dimethoxy-phenyl)-6,6-dimethyl-8-oxo-4,5,6,7,8,9-hexahydro- pyrazolo[5,l-b]quinazoline-3-carboxylic acid 4-nitro-phenyl ester (59.7 mg, 0.1 mmol) was taken in dry DMF (1 ml) under argon and charged with thiophene-2-carboxylic acid hydrazide (42.6 mg, 0.3 mmol) and stirred at 60 C till the reation was complete.
The reaction mixture was diluted with 1: 1 mixture of DMSO/CAN to 2 ml, filter through a microfilter and the product was purified by HPLC to yield 21 mg of the pure product. Example 26 9-(3-Bromo-4,5-dimethoxy-phenyl)-6,6-dimethyl-8-oxo-4,5,6,7,8,9-hexahy dro-pyrazolo[5,l-b]quinazoline-3-carboxylic acid {3-[4-(3,5-dichloro-phenyl)- piperazin- 1 -yl] - 1 -methy 1-propoxy }-amide
Figure imgf000061_0002
9-(3-Bromo-4,5-dimethoxy-phenyl)-6,6-dimethyl-8-oxo-4,5,6,7,8,9-hexahydro- pyrazolo[5,l-b]quinazoline-3-carboxylic acid 4-nitro-phenyl ester (59.7 mg, 0.1 mmol) was taken in dry DMF (1 ml) under argon and charged with 0-{3-[4-(3,5-Dichloro- phenyl)-piperazin-l-yl] -propyl} -hydroxylamine dihydrochloride (112.8 mg, 0.3 mmol) and stirred at 60 C till the reation was complete.
The reaction mixture was diluted with 1: 1 mixture of DMSO/CAN to 2 ml, filter through a microfilter and the product was purified by HPLC to yield 44 mg of the pure product.
Example 27 9-(3-Bromo-4,5-dimethoxy-phenyl)-6,6-dimethyl-8-oxo-4,5,6,7,8,9-hexahy dro-pyrazolo[5,l-b]quinazoline-3-carboxylic acid N'-(2-lH-indol-3-yl-acetyl)- hydrazide
Figure imgf000062_0001
9-(3-Bromo-4,5-dimethoxy-phenyl)-6,6-dimethyl-8-oxo-4,5,6,7,8,9-hexahydro- pyrazolo[5,l-b]quinazoline-3-carboxylic acid 4-nitro-phenyl ester (59.7 mg, 0.1 mmol) was taken in dry DMF (1 ml) under argon and charged with lH-Indole-3-carboxylic acid hydrazide (52.5 mg, 0.3 mmol) and stirred at 60 C till the reation was complete.
The reaction mixture was diluted with 1: 1 mixture of DMSO/CAN to 2 ml, filter through a microfilter and the product was purified by HPLC to yield 54 mg of the pure product. Example 28 4-Methyl-[l,2,3]thiadiazole-5-carboxylic acid N'-[9-(3-bromo-4,5-dimet hoxy^henyl)-6,6-dimethyl-8-oxo-4,5,6,7,8,9-hexahydro-pyrazolo[5,l-b]quinazoline-3- carbonyl] -hydrazide
Figure imgf000063_0001
9-(3-Bromo-4,5-dimethoxy-phenyl)-6,6-dimethyl-8-oxo-4,5,6,7,8,9-hexahydro- pyrazolo[5,l-b]quinazoline-3-carboxylic acid 4-nitro-phenyl ester (59.7 mg, 0.1 mmol) was taken in dry DMF (1 ml) under argon and charged with 4-Methyl-[l,2,3]thiadiazole-5- carboxylic acid hydrazide (47.4 mg, 0.3 mmol) and stirred at 60 C till the reation was complete.
The reaction mixture was diluted with 1: 1 mixture of DMSO/CAN to 2 ml, filter through a microfilter and the product was purified by HPLC to yield 35 mg of the pure product. Example 29: Examination of Wnt signaling pathway inhibiting activity of selected compounds
To screen for small-molecule modulators of the Wnt signaling pathway, a reporter gene based assay describing the modulation of the TCF4 transcription factor was used. More specifically 4000 Hek293T cells were seeded into 384 high density plates. 24h after seeding a Wnt-sensitive reporter (6xTCF-luciferase (Firefly) (pTOP-FLASH; "Armadillo coactivates transcription driven by the product of the Drosophila segment polarity gene dTCF", Cell, 1997, 88(6), pages 789-99)) and constitutively expressed control reporter (Renilla luciferase pCMV-RL) were transfected into Hek293T. Wnt signaling was stimulated by cotransfecting mouse Wntl, mouse Frizzled 8 and human LRP6 according to "Casein kinase 1 gamma couples Wnt receptor activation to cytoplasmic signal transduction", Nature, 2005, 438(7069), pages 867-872. 24 h after pathway stimulation compounds were added at a concentration of 10 microM and allowed to incubate for 24 h. For the evaluation of the IC50 the respective compound was applied in increasing concentrations yielding final concentrations per well of 5nM - 100 microM.
The IC5o-values of the Wnt pathway inhibitory activity of compounds (1), (4), (6), (7), (16) to (20), (29) to (32) and (39) to (43) are shown in table 1.
Table 1: I C50- values of the Wnt pathway inhibitory activity for of compounds (23) to (28)
Figure imgf000064_0001
Example 30: Examination of toxicity in Hek293T and HepG2
For examination of cytotoxicity in these cancer cell lines the commercial available CellTiterGlo Reagent (Promega, USA) was used according to the manufactor.
Compounds were applied to Hek293T (denoted (a)) or HepG2 (denoted (b)), cultured in Dulbecco's Modified Eagles Medium, supplemented with 10% fetal calf serum and 1% penicillin/streptomycin. Cells were grown in T75 flasks at 37 °C, 5% C02 and trypsinized when 60-80% confluent by adding 2 ml of 0.25% Trypsin-EDTA solution. Cells were then re-suspended into culture medium yielding approx. 4000 cells (a) or 4500 cells (b), suspended in 30 microl medium. 48h after cell seeding, compounds were added to yield the desired final concentrations. 24h after compound addition cytotoxicity was evaluated. For this purpose the media was removed and CellTiterGlo was added according to the manufactors manual. In this assay the luciferase emission readout is directly correlated with the cellular amount of ATP, low luciferase emission thus is reflecting cytotoxicity of a compound. For the evaluation of the IC50 the compound was applied in increasing concentrations yielding final concentrations per well of 5nM - 100 microM.
IC5o-values of the cytotoxic activity of compounds (23) to (28) against Hek293T and HepG2 are shown in table 2. Table 2: IC50-values against Hek293T and HepG2 of compounds (23) to (28)
Figure imgf000065_0001
Example 31: Examination of cell line specific cytotoxicity
For examination of cell line- specific cytotoxicity, compounds were applied to human colorectal cancer cells (HCT116, denoted (1); SW480, denoted (2); Dld-1, denoted (3)) and human fibroblasts (HS-68, denoted (4)), cultured in Mc Coy's ((1)) and Dulbecco's Modified Eagles Medium ((2)-(4)), supplemented with 10% ((l)-(3)) and 20% ((4)) fetal calf serum and 1% penicillin/streptomycin. Cells were grown in T75 flasks at 37 °C, 5% C02 and trypsinized when 60-80% confluent by adding 2 ml of 0.25% Trypsin-EDTA (ethylenediaminetetraacetic acid) solution. Cells were then re-suspended into culture medium yielding approx. 750 cells, suspended in 45 microl medium, were plated into each well of a black 384-well plate for fluorescence imaging experiments. 24 - 36 hr later 5 microl compound solution (100 microM compound dissolved in ultra pure water containing 1% DMSO (dimethylsulf oxide)), were added to achieve a final concentration of 10 microM and were incubated for at least 72 hr. Compound incubation was terminated by (a) fixation and (b) permeabilisation of cells, followed by (c) fluorescence labeling of cell nuclei for cytometric quantification or by immunocytochemistry for microscopic evaluation of cell morphology. The three steps were performed by replacing the solution of the previous step in each well of a 384-well plate by (a) 30 microl PBS (phosphate buffered saline) containing 5% PFA (paraformaldehyde), (b) 30 microl PBS containing 0.2% TritonX-100 and (c) 10 microl PBS containing Hoechst-33342 for cytometry or Hoechst-33342, FITC- (fluorescein isothiocyanate-)labeled alpha-tubulin antibodies and TRrrC-(tetramethylrhodamine isothiocyanate-)labeled phalloidin for microscopy. Solutions were incubated for 15 (a,b) and 30 min (c) in the dark at room temperature. Cells were washed twice between each step with 30 microl PBS. The assay system was miniaturized and adapted to automated workflow using liquid-handling robotics. Cell line- specific cytotoxicity was quantified by counting the number of Hoechst labeled nuclei per well of a 384- well plate using a plate cytometer. Data obtained with the cytometer was analysed using standard data analysis software. Identified hits were further evaluated by visual inspection of fluorescence micrographs obtained from imaging using a conventional fluorescence microscope.
The results for normal cells (HS68) are shown in table 3. Table 3: Influence of compounds (23) to (28) on normal cells (HS68), Dldl, HCT116 and SW480 colon carcinoma cells
Figure imgf000066_0001
The examination of the antiproliferative activity against human colon carcinoma cells Dldl, HCT116 and SW480 was carried out in analogy to the procedure carried out for HS68. The respective compound applied in increasing concentrations yielding final concentrations per well of 30nM - 100 microM. Compounds of the present invention have potent antiproliferative acivity against human colon carcinoma cells as shown for Dldl, HCT116 and SW480 in Table 3, too.

Claims

Claims
1. A compound of formula (I)
Figure imgf000067_0001
(I)
wherein
X1 is CR1 or N;
X2 is CR2 or N;
X3 is CR3 or N;
with at least one of the group X 1 ; X2 ; and X 3 is N;
Z is CR4 or CR4R5;
Y is N or NR6;
the dotted line denotes 1 or 2 double bonds; L is O or NR7;
each A is independently from each other CR 8 R 8<i which is same or different and one A is optionally selected from the group consisting of O; S; NR9; S(O) and S(0)2;
n is 2 to 4;
R 1^ R2"; and R 3J are independently from each other selected from H; OH; halogen;
CN; Ci-Ce alkyl; C3-C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl; C4-C15 aralkyl; C4-C15 heteroarylalkyl; ORla;
C(0)Rla; C(0)ORla; C(0)NRlaRlb; C(0)N(Rla)ORlb;
C(0)N(Rla)N(Rlb)C(0)Rlc; C(0)N(Rla)N(Rlb)Rlc; S(0)2N(RlaRlb);
S(0)N(RlaRlb); S(0)2Rla; S(0)Rla; N(Rla)S(0)2N(RlbRlc); SRla; OC(0)Rla; N(Rla)C(0)Rlb; N(Rla)S(0)2Rlb; N(Rla)S(0)Rlb; N(Rla)C(0)N(RlbRlc); N(Rla)C(0)ORlb; OC(0)N(RlaRlb); CHN(Rla); and CHNN(RlaRlb) wherein alkyl; cycloalkyl; heterocyclyl; aryl; heteroaryl; aralkyl; and heteroarylalkyl are optionally substituted by one or more groups R which are same or different;
Rla; Rlb; and Rlc are independently from each other selected from H; CrC6 alkyl; C3- C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl; C8-C12 biaryl; C8-C12 heterobiaryl; C4-C15 aralkyl; and C4-C15 heteroarylalkyl; wherein alkyl; cycloalkyl; heterocyclyl; alkenyl; aryl;
biaryl; heteroaryl; heterobiaryl; aralkyl; and heteroarylalkyl are optionally substituted with one or more R10 which are the same or different ;
R10 is selected from halogen, CN, OH, C1-6 alkyl; C3-C7 cyclyl; C3-C7
heterocyclyl; C3-C7 aryl; C3-C7 heteroaryl; C8-C12 biaryl; C8-C12
heterobiaryl; OR10a; C(O)R10a; C(O)OR10a; NR10aR10b; C(O)NR10aR10b; S(O)2N(R10aR10b); S(O)N(R10aR10b); S(O)2R10a; S(O)R10a;
N(R10a)S(O)2N(R10bR10c); SR10a; OC(O)R10b; N(R10a)C(O)R10b;
N(R10a)S(O)2R10b; N(R10a)S(O)R10b; N(R10a)C(O)N(R10bR10c);
N(R10a)C(O)OR10b; and OC(O)N(R10aR10b); wherein cyclyl; heterocyclyl; aryl; heteroaryl; biaryl; heterobiaryl; and alkyl are optionally substituted with one or more R10a, which are the same or different;
R10a; R10b and R10c are independently from each other selected from H; halogen; C3- C7 cyclyl; C3-C7 heterocyclyl; C3-C7 aryl; C3-C7 heteroaryl; and CrC6 alkyl; wherein cyclyl; heterocyclyl; aryl; heteroaryl and alkyl are optionally substituted with one or more halogen which are the same or different;
R4 is a 5 to 6 membered aromatic or hetero aromatic ring which is optionally
substituted by one or more R4a;
R4a is selected from H; halogen, CN, OH, OR4b, C(0)R4b; C(0)OR4b; NR4bR4c;
C(0)NR4bR4c; S(0)2N(R4bR4c); S(0)N(R4bR4c); S(0)2R4b; S(0)R4b;
N(R4b)S(0)2N(R4cR4d); SR4b; OC(0)R4b; N(R4b)C(0)R4c; N(R4b)S(0)2R4c; N(R4b)S(0)R4c; N(R4b)C(0)N(R4cR4d); N(R4b)C(0)OR4c; OC(0)N(R4b R4c); and C C6 alkyl; wherein alkyl is optionally substituted with one or more R 13 which are the same or different ; R1J is selected from halogen, CN, OH, C1-6 alkyl; OR1Ja; C(0)R1Ja; C(0)OR1Ja;
NRi3aRi3b. C(0)NR13aR13b; S(0)2N(R13aR13b); S(0)N(R13aR13b); S(0)2R13a; S(0)R13a; N(R13a)S(0)2N(R13bR13c); SR13a; OC(0)R13a; N(R13a)C(0)R13b; N(R13a)S(0)2R13b; N(R13a)S(0)R13b; N(R13a)C(0)N(R13bR13c);
N(R13a)C(0)OR13b; and OC(0)N(R13a R13b); wherein C1-6 alkyl is optionally substituted with one or more halogen, which are the same or different;
R4b, R4c, R4d, R13a; R13b and R13c are independently from each other selected from H; and C C6 alkyl; wherein Ci_6 alkyl is optionally substituted with one or more halogen, which are the same or different;
R5 is H or linear C C6 alkyl;
R6 is selected from H; OH; C C6 alkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl;C4-C15 aralkyl; C4-C15 heteroarylalkyl; OR6a;
C(0)R6a; C(0)OR6a; OC(0)R6a; OC(0)N(R6aR6b); N(R6aR6b);
N(R6a)C(0)N(R6bR6c); and N(R6a)C(0)OR6b wherein alkyl; cycloalkyl; heterocyclyl; alkenyl; aryl; heteroaryl; aralkyl; and heteroarylalkyl are optionally substituted by one ore more R11, which are the same or different;
R6a; R6b; and R6c are independently from each other selected from H; Q-C6 alkyl; C3- C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl; C4-C15 aralkyl; and C4-C15 heteroarylalkyl which are optionally substituted with one or more R11, which are the same or different;
R11 is selected from C1-6 alkyl; halogen, CN; OH; ORl la; C(0)Rl la; C(0)ORlla;
NRi iaRi ib. C(0)NRllaRllb; S(0)2N(RllaRllb); S(0)N(RllaRllb); S(0)2Rlla; S(0)Rlla; N(Rlla)S(0)2N(Rl lbRllc); SRlla; OC(0)Rllb; N(Rlla)C(0)Rllb; N(Rl la)S(0)2Rlla; N(Rlla)S(0)Rl lb; N(Rlla)C(0)N(RllbRl lc);
N(Rl la)C(0)ORl lb; and OC(0)N(RllaRllb); wherein C1-6 alkyl is optionally substituted with one or more halogen, which are the same or different;
Ri ia. Rnb. an(j Ri ic ^ mcjepencientiy fr0m each other selected from H; and C C6 alkyl; wherein Ci_6 alkyl is optionally substituted with one or more halogen, which are the same or different; selected from H; OH; CrC6 alkyl; C3-C7 cycloalkyl; C3-C7 heterocyclyl; C2 C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl; C4-C15 aralkyl; C4-C15
heteroarylalkyl; OR7a; C(0)R7a; C(0)OR7a; OC(0)R7a; OC(0)N(R7aR7b); N(R7aR7b); N(R7a)C(0)R7b; N(R7a)C(0)N(R7bR7c); and N(R7a)C(0)OR7b; wherein alkyl; cycloalkyl; alkenyl; aryl; aralkyl; and heteroarylalkyl are
12
optionally substituted by one ore more R , which are the same or different;
; and R7c are independently from each other selected from H; CrC6 alkyl; C3 C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl; C4-C15 aralkyl; and C4-Q5 heteroarylalkyl wherein alkyl;
cycloalkyl; heterocyclyl; alkenyl; aryl; heteroaryl; aralkyl; heteroarylalkyl
12
are optionally substituted with one or more R , which are the same or different ;
R12 is selected from C1-6 alkyl; halogen, CN, OH, OR12a; C(0)R12a; C(0)OR12a;
NRi2aRi2b. C(0)NR12aR12b; S(0)2N(R12aR12b); S(0)N(R12aR12b); S(0)2R12a; S(0)R12a; N(R12a)S(0)2N(R12bR12c); SR12a; OC(0)R12b; N(R12a)C(0)R12b; N(R12a)S(0)2R12a; N(R12a)S(0)R12b; N(R12a)C(0)N(R12bR12c);
N(R12a)C(0)OR12b; and OC(0)N(R12aR12b); wherein C1-6 alkyl is optionally substituted with one or more halogen, which are the same or different;
R 12 a; R 12b 12
and R c are independently from each other selected from H; and C C6 alkyl; wherein C1-6 alkyl is optionally substituted with one or more halogen; which are the same or different;
R ; and R a are independently from each other selected from H and C¾;
R9 is selected from OH; C C6 alkyl; C3-C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3-C7 aryl; C3-C7 heteroaryl; C4-C15 aralkyl; C4-C15
heteroarylalkyl; OR9a; C(0)R9a and C(0)OR9a; wherein alkyl; cycloalkyl; alkenyl; aryl; aralkyl; and heteroarylalkyl are optionally substituted by one ore more groups selected from OH; halogen; CN; CrC6 alkyl; OR9b;
C(0)R9b; C(0)OR9b; NR9bR9c; C(0)NR9bR9c; S(0)2N(R9bR9c);
S(0)N(R9bR9c); S(0)2R9b; S(O) R9b; N(R9b)S(0)2N(R9cR9d); SR9b;
OC(0)R9b; N(R9b)C(0)R9c; N(R9b)S(0)2R9c; N(R9b)S(0)R9c;
N(R9b)C(0)N(R9cR9d); N(R9b)C(0)OR9c; and OC(0)N(R9bR9c); wherein C1-6 alkyl is optionally substituted with one or more halogen, which are the same or different;
R a; R and R c are independently from each other selected from H; and CrC6 alkyl; wherein C1-6 alkyl is optionally substituted with one or more halogen; which are the same or different; and/or solvates; hydrates; and pharmaceutically acceptable salts thereof.
The compound according to claim 1, wherein R4 is selected from C6 aryl and C 6 heteroaryl and is substituted in p- and both m-positions by three substituents selected iinnddeeppeennddeennttllyy ffrroomm eeaacchh ootthheerr ffrroonm R4a and/or solvates; hydrates and pharmaceutically acceptable salts thereof.
3. The compound according to claim 1 or 2, wherein X1 is CR1 with R1 selected from OH; halogen; CrC6 alkyl; C3-C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl; C3- C7 heteroaryl; C4-C15 aralkyl; C4-C15 heteroarylalkyl; ORla; C(0)Rla;
C(0)N(Rla)ORlb; C(0)N(Rla)N(Rlb)C(0)Rlc; C(0)N(Rla)N(Rlb)Rlc;
S(0)2N(RlaRlb); S(0)N(RlaRlb); S(0)2Rla; S(0)Rla; N(Rla)S(0)2N(RlbRlc); SRla; OC(0)Rla; N(Rla)S(0)2Rlb; N(Rla)S(0)Rlb; and N(Rla)C(0)N(RlbRlc)
and/or solvates; hydrates; and pharmaceutically acceptable salts thereof.
4. The compound according to claim 1, wherein at least one of XI, X2 and X3 is not N and the respective group R1; R2 or R3 is selected from C(0)N(Rla)ORlb;
C(0)N(Rla)N(Rlb)Rlc; and C(0)N(Rla)N(Rlb)C(0)Rlc
and/or solvates; hydrates; and pharmaceutically acceptable salts thereof.
5. The compound according to any of claim 1 wherein R4 is selected from C6 aryl and C6 heteroaryl and is substituted in p- and both m-positions by three substituents selected independently from each other from R4a and X1 is CR1 with R1 selected from
OH; halogen; CN; CrC6 alkyl; C3-C7 cycloalkyl; C3-C7 heterocyclyl; C2-C6 alkenyl;
C3-C7 aryl; C3-C7 heteroaryl; C4-C15 aralkyl; C4-C15 heteroarylalkyl; ORla; C(0)Rla;
C(0)ORla; C(0)N(RlaRlb); C(0)N(Rla)ORlb; C(0)N(Rla)N(Rlb)C(0)Rlc;
C(0)N(Rla)N(Rlb)Rlc; S(0)2N(RlaRlb); S(0)N(RlaRlb); S(0)2Rla; S(0)Rla;
N(Rla)S(0)2N(RlbRlc); SRla; OC(0)Rla; N(Rla)C(0)Rlb; N(Rla)S(0)2Rlb;
N(Rla)S(0)Rlb; and N(Rla)C(0)N(RlbRlc) and/or solvates; hydrates; and pharmaceutically acceptable salts thereof.
6. The compound according to claim 1 wherein R4 is selected from C6 aryl and C6
heteroaryl and is substituted in p- and both m-positions by three substituents selected independently from each other from R4a and wherein at least one of XI, X2 and X3 is not N and the respective group R1; R2 or R3 is selected from C(0)N(Rla)ORlb; C(0)N(Rla)N(Rlb)Rlc; and C(0)N(Rla)N(Rlb)C(0)Rlc
and/or solvates; hydrates; and pharmaceutically acceptable salts thereof.
7. A compound according to any of claims 1 to 6 for use as medicament.
8. Use of the compound according to any of claims 1 to 6 for modulating the Wnt signalling pathway.
9. A compound according to any of claims 1 to 6 for modulating the Wnt signalling pathway.
10. A compound according to any of claims 1 to 6 for the treatment of a disorder or disease associated with an aberrant activation of Wnt signalling in a mammal.
11. The compound according to claim 10 wherein the Wnt associated disorder or disease is cancer, a proliferative skin disorder, rheumatoid arthritis, increased bone density, aging or age-related disorders and/or diseases or Dupuytren disease (superficial fibromatosis).
12. The compound according to claim 11 wherein the cancer is member of the group multiple myeloma, colon cancer, breast cancer, gastritic cancer, colorectal cancer, lung cancer, prostate cancer, ovarian cancer, bladder cancer, liver cancer, uterine cancer, kidney cancer, leukaemia, gliomas, basal cell carcinoma, rhabdomyosarcoma, mesothelioma, osteosarcoma, medulloblastomas and other primary CNS malignant neuroectodermal tumors.
13. A pharmaceutical composition containing a compound of any of claims 1 to 6.
14. Use of the compound according to any of claims 1 to 6 in a kit.
15. Method for the preparation of a medicament comprising the steps of:
a) preparing at least one compound according to any of claims 1 to 6; and b) formulating a medicament containing at least said compound.
16. Method of treating a mammal for the modulation of the Wnt signalling pathway, which method comprises administering to said mammal a therapeutically effective amount of a compound according to any of claims 1 to 6.
17. A process for the preparation of a compound according to any of claims 1 to 6 comprising reacting an aldehyde R4-CHO, a 5-to-7-membered cyclic 1,3-diketone and a heteroaromate substituted by NH2 wherein R4 is defined as in formula (I).
PCT/EP2011/055060 2010-03-31 2011-03-31 PYRAZOL[1,5-a]PYRIMIDINE DERIVATIVES AS WNT PATHWAY ANTAGONISTS WO2011121096A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP10158764.0 2010-03-31
EP10158764 2010-03-31

Publications (1)

Publication Number Publication Date
WO2011121096A1 true WO2011121096A1 (en) 2011-10-06

Family

ID=42320600

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/055060 WO2011121096A1 (en) 2010-03-31 2011-03-31 PYRAZOL[1,5-a]PYRIMIDINE DERIVATIVES AS WNT PATHWAY ANTAGONISTS

Country Status (1)

Country Link
WO (1) WO2011121096A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8957078B2 (en) 2013-03-15 2015-02-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8969360B2 (en) 2013-03-15 2015-03-03 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9309250B2 (en) 2011-06-22 2016-04-12 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-b]pyrazines as ATR kinase inhibitors
US9340546B2 (en) 2012-12-07 2016-05-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
JP2016540803A (en) * 2013-12-17 2016-12-28 エージェンシー フォー サイエンス,テクノロジー アンド リサーチ WNT pathway modulator
US9663519B2 (en) 2013-03-15 2017-05-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9670215B2 (en) 2014-06-05 2017-06-06 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10160760B2 (en) 2013-12-06 2018-12-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
CN113354651A (en) * 2020-07-30 2021-09-07 四川大学 Pyrazolo [1,5-a ] quinazoline derivative and application thereof in preparation of medicines
US11179394B2 (en) 2014-06-17 2021-11-23 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of Chk1 and ATR inhibitors
US11464774B2 (en) 2015-09-30 2022-10-11 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA damaging agents and ATR inhibitors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5041138A (en) 1986-11-20 1991-08-20 Massachusetts Institute Of Technology Neomorphogenesis of cartilage in vivo from cell culture
WO2001036422A1 (en) * 1999-11-19 2001-05-25 Abbott Laboratories Tricyclic dihydropyrimidine potassium channel openers
US20070219257A1 (en) 2004-09-29 2007-09-20 Beachy Philip A Wnt Pathway Antagonists

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5041138A (en) 1986-11-20 1991-08-20 Massachusetts Institute Of Technology Neomorphogenesis of cartilage in vivo from cell culture
WO2001036422A1 (en) * 1999-11-19 2001-05-25 Abbott Laboratories Tricyclic dihydropyrimidine potassium channel openers
US20070219257A1 (en) 2004-09-29 2007-09-20 Beachy Philip A Wnt Pathway Antagonists

Non-Patent Citations (45)

* Cited by examiner, † Cited by third party
Title
"Armadillo coactivates transcription driven by the product of the Drosophila segment polarity gene dTCF", CELL, vol. 88, no. 6, 1997, pages 789 - 99
"Casein kinase 1 gamma couples Wnt receptor activation to cytoplasmic signal transduction", NATURE, vol. 438, no. 7069, 2005, pages 867 - 872
ABOUL-FETOUH E MOURAD ET AL., BEILSTEIN J ORG CHEM, vol. 3, no. 11, 2007, pages 1-5, XP002592765 *
APELQVIST ET AL., CURR. BIOL., vol. 7, 1997, pages 801 - 4
BARKER NICK ET AL: "Mining the Wnt pathway for cancer therapeutics", NATURE REVIEWS. DRUG DISCOVERY, NATURE PUBLISHING GROUP, GB LNKD- DOI:10.1038/NRD2154, vol. 5, no. 12, 1 December 2006 (2006-12-01), pages 997 - 1014, XP002545380, ISSN: 1474-1784 *
BELLUSCI ET AL., DEVELOPMENT, vol. 124, 1997, pages 53
BUCHWALD ET AL., SURGERY, vol. 88, 1980, pages 507
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 October 2010 (2010-10-12), XP002639462, retrieved from STN Database accession no. 1246072-53-6 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 October 2010 (2010-10-12), XP002639463, retrieved from STN Database accession no. 1246055-25-3 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 October 2010 (2010-10-12), XP002639464, retrieved from STN Database accession no. 1246044-15-4 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 October 2010 (2010-10-12), XP002639466, retrieved from STN Database accession no. 1246071-96-4 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 October 2010 (2010-10-12), XP002639467, retrieved from STN Database accession no. 1246069-59-9 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 October 2010 (2010-10-12), XP002639468, retrieved from STN Database accession no. 1246069-49-7 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 19 April 2010 (2010-04-19), XP002639465, retrieved from STN Database accession no. 1219561-43-9 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 26 September 2007 (2007-09-26), XP002592764, retrieved from STN Database accession no. 948141-77-3 *
DRIZIN I ET AL, BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, PERGAMON, ELSEVIER SCIENCE, GB LNKD- DOI:10.1016/S0960-894X(02)00207-X, vol. 12, 1 January 2002 (2002-01-01), pages 1481 - 1484, XP002362324, ISSN: 0960-894X *
DURING ET AL., ANN. NEUROL., vol. 25, 1989, pages 351
EL-GAZZAR A R B A ET AL, EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, EDITIONS SCIENTIFIQUE ELSEVIER, PARIS, FR, vol. 44, no. 10, 1 October 2009 (2009-10-01), pages 4249 - 4258, XP026394776, ISSN: 0223-5234, [retrieved on 20090528] *
FREED ET AL., J. BIOMED. MATER. RES., vol. 27, 1993, pages 11
FUJITA ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 238, 1997, pages 658
GOODRICH ET AL., SCIENCE, vol. 277, 1997, pages 1109 - 13
GOODSON, MEDICAL APPLICATIONS OF CONTROLLED RELEASE, vol. 2, 1984, pages 115
GRANDE ET AL., J. ORTHOP. RES., vol. 7, 1989, pages 208
HOWARD ET AL., J. NEUROSURG., vol. 71, 1989, pages 858
HUANG ET AL., NATURE, vol. 461, 2009, pages 614 - 620
JOHNSON ET AL., SCIENCE, vol. 272, 1996, pages 1668 - 71
LANGER, SCIENCE, vol. 249, 1990, pages 1527
LEVY ET AL., SCIENCE, vol. 228, 1985, pages 190
LI ET AL., PNAS, vol. 99, 2002, pages 13254 - 13259
LU ET AL., BMC PHARM, vol. 9, 2009, pages 13
MAJID M HERAVI ET AL, MOLECULAR DIVERSITY, KLUWER ACADEMIC PUBLISHERS, DO LNKD- DOI:10.1007/S11030-008-9086-8, vol. 12, no. 3-4, 9 September 2008 (2008-09-09), pages 181 - 185, XP019647073, ISSN: 1573-501X *
N. BARKER, H. CLEVERS: "Mining the Wnt pathway for cancer therapeutics", NATURE REVIEWS, vol. 5, 2007, pages 997 - 1014
ORO ET AL., SCIENCE, vol. 276, 1997, pages 817 - 21
R. NUSSE: "Wnt signalling in disease and in development", CELL RESEARCH, vol. 15, 2005, pages 23 - 32
RANGER, PEPPAS, MACROMOL. SCI. REV. MACROMOL. CHEM., vol. 23, 1983, pages 61
SAUDEK ET AL., N. ENGL. J. MED., vol. 321, 1989, pages 574
SCHROEDER ET AL., J. BIOMED. MATER. RES., vol. 25, 1991, pages 329
SEFTON, CRC CRIT. REF. BIOMED. ENG., vol. 14, 1987, pages 201
STONE ET AL., CLIN. ORTHOP. RELAT. RED, vol. 252, 1990, pages 129
STONE ET AL., NATURE, vol. 384, 1996, pages 129 - 34
TAKIGAWA ET AL., BONE MINER, vol. 2, 1987, pages 449
VACANTI ET AL., PLAST. RECONSTR. SURG., vol. 88, 1991, pages 753
WALITANI ET AL., J. BONE JT. SURG., vol. 71B, 1989, pages 74
XIE ET AL., CANCER RES., vol. 57, 1997, pages 2369 - 72
XIE ET AL., GENES CHROMOSOMES CANCER, vol. 18, 1997, pages 305 - 9

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9309250B2 (en) 2011-06-22 2016-04-12 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-b]pyrazines as ATR kinase inhibitors
US10392391B2 (en) 2012-12-07 2019-08-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9340546B2 (en) 2012-12-07 2016-05-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11370798B2 (en) 2012-12-07 2022-06-28 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9650381B2 (en) 2012-12-07 2017-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11117900B2 (en) 2012-12-07 2021-09-14 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9718827B2 (en) 2012-12-07 2017-08-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10787452B2 (en) 2012-12-07 2020-09-29 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8969360B2 (en) 2013-03-15 2015-03-03 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9663519B2 (en) 2013-03-15 2017-05-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8957078B2 (en) 2013-03-15 2015-02-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10160760B2 (en) 2013-12-06 2018-12-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10815239B2 (en) 2013-12-06 2020-10-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11485739B2 (en) 2013-12-06 2022-11-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
JP2016540803A (en) * 2013-12-17 2016-12-28 エージェンシー フォー サイエンス,テクノロジー アンド リサーチ WNT pathway modulator
US10093676B2 (en) 2014-06-05 2018-10-09 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10800781B2 (en) 2014-06-05 2020-10-13 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9670215B2 (en) 2014-06-05 2017-06-06 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11179394B2 (en) 2014-06-17 2021-11-23 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of Chk1 and ATR inhibitors
US11464774B2 (en) 2015-09-30 2022-10-11 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA damaging agents and ATR inhibitors
CN113354651A (en) * 2020-07-30 2021-09-07 四川大学 Pyrazolo [1,5-a ] quinazoline derivative and application thereof in preparation of medicines
CN113354651B (en) * 2020-07-30 2022-07-22 四川大学 Pyrazolo [1,5-a ] quinazoline derivative and application thereof in preparation of medicines

Similar Documents

Publication Publication Date Title
WO2011121096A1 (en) PYRAZOL[1,5-a]PYRIMIDINE DERIVATIVES AS WNT PATHWAY ANTAGONISTS
US10065939B2 (en) Chromene derivatives and their analogs as Wnt pathway antagonists
WO2010149786A1 (en) Pyrido [2, 3-d] pyrimidines as wnt antagonists for treatment of cancer and arthritis
AU780846B2 (en) Mediators of hedgehog signaling pathways, compositions and uses related thereto
AU2002327390B2 (en) Mediators of hedgehog signaling pathways, compositions and uses related thereto
US9173869B2 (en) Mediators of hedgehog signaling pathways, compositions and uses related thereto
JP2005529876A (en) Modulators of the Hedgehog signaling pathway, compositions related thereto and uses
US8629272B2 (en) Tricyclic pyrimidine derivatives as wnt antagonists
AU2011224045A1 (en) Modulators of hedgehog signaling pathways, compositions and uses related thereto

Legal Events

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

Ref document number: 11716191

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11716191

Country of ref document: EP

Kind code of ref document: A1