WO2017055313A1 - Amido-substituted azole compounds - Google Patents

Amido-substituted azole compounds Download PDF

Info

Publication number
WO2017055313A1
WO2017055313A1 PCT/EP2016/073040 EP2016073040W WO2017055313A1 WO 2017055313 A1 WO2017055313 A1 WO 2017055313A1 EP 2016073040 W EP2016073040 W EP 2016073040W WO 2017055313 A1 WO2017055313 A1 WO 2017055313A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
general formula
carbamoyl
phenyl
Prior art date
Application number
PCT/EP2016/073040
Other languages
French (fr)
Inventor
Knut Eis
Jens Ackerstaff
Sarah WAGNER
Philipp BUCHGRABER
Detlev Sülzle
Eckhard Bender
Volkhart Min-Jian Li
Ningshu Liu
Franziska SIEGEL
Philip Lienau
Original Assignee
Bayer Pharma Aktiengesellschaft
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 Bayer Pharma Aktiengesellschaft filed Critical Bayer Pharma Aktiengesellschaft
Publication of WO2017055313A1 publication Critical patent/WO2017055313A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to amido-substituted azole compounds of general formula (I) as described and defined herein, to methods of preparing said compounds, to intermediate compounds useful for preparing said compounds, to pharmaceutical compositions and combinations comprising said compounds and to the use of said compounds for manufacturing a pharmaceutical composition for the treatment or prophylaxis of a disease, in particular of neoplasms, as a sole agent or in combination with other active ingredients.
  • Cancer is the leading cause of death in developed countries and the second leading cause of death in developing countries. Deaths from cancer worldwide are projected to continue rising, with an estimated 12 million deaths in 2030. While substantial progress has been made in developing effective therapies, there is a need for additional therapeutic modalities that target cancer and related diseases.
  • cancer stem cells represent the apex in the hierarchical model of tumor genesis, heterogeneity and metastasis.
  • CSCs possess the capacity for unlimited self-renewal, the ability to give rise to progeny cells, and also an innate resistance to cytotoxic therapeutics [Meacham CE and Morrison SJ. Tumour heterogeneity and cancer cell plasticity. Nature 2013 , 501 :328]. Thus, there is need to develop drugs for cancer therapy addressing distinct features of established tumors.
  • Wnt signaling cascades have classified into two categories: canonical and non- canonical, differentiated by their dependence on ⁇ -catenin.
  • Non-canonical Wnt pathways such as the planar cell polarity (PCP) and Ca 2+ pathway, function through ⁇ -catenin independent mechanisms.
  • Canonical Wnt signalling is initiated when a Wnt ligand engages co- receptors of the Frizzled (Fzd) and low- density lipoprotein receptor related protein (LRP) families, ultimately leading to ⁇ -catenin stabilization, nuclear translocation and activation of target genes [Angers S, Moon RT. Proximal events in Wnt signal transduction. Nat Rev Mol Cell Biol. 2009, 10: 468.
  • ⁇ -catenin In the absence of Wnt stimulus, ⁇ -catenin is held i n an inactive state by a multimeric "destruction" complex comprised of adenomatous polyposis coli (APC), Axin , glycogen synthase kinase 36 (GSK36) and casein kinase 1 a (CK1 a).
  • APC and Axin function as a scaffold, permitting GSK36- and CK1 a-mediated phosphorylation of critical residues within ⁇ -catenin.
  • ⁇ -catenin is stabilized and translocated to the nucleus.
  • 6- catenin forms a complex with members of the T-cell factor/lymphoid enhancer factor (TCF/ LEF) family of transcription factors, recruiting co-factors such as CBP, p300, TNIK, Bcl9 and Pygopus, and ultimately driving transcription of target genes including c-myc, 0ct4, cyclin D, survivin.
  • TCF/ LEF T-cell factor/lymphoid enhancer factor
  • Tankyrases play a key role in the destruction complex by regulating the stability of the rate-limiting AXIN proteins, RNF1 6 and tankyrase itself.
  • the E3 ubiquitin ligase RNF1 6 recognizes tankyrase-mediated PARsylation and eartags AXIN, tankyrase and itself for proteasome-mediated degradation.
  • tankyrases control the protein stability and turnover of key components of the destruction complex, and consequently the cellular levels of ⁇ -catenin [Huang SMA, Mishina YM, Liu S, Cheung A, Stegmeier F, et al.
  • Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature 2009, 461:614, Zhang Y, Liu S, Mickanin C, Feng Y, Charlat 0, et al.
  • RNF146 is a poly(ADP-ribose)-directed E3 ligase that regulates axin degradation and Wnt signalling. Nature Cell Biology 2011, 13:623, 2011].
  • Wnt/B-catenin signaling pathway Aberrant regulation of the Wnt/B-catenin signaling pathway is a common feature across a broad spectrum of human cancers and evolves as a central mechanism in cancer biology.
  • Wnt overexpression could lead to malignant transformation of mouse mammary tissue [Klaus A, BirchmeierW. Wnt signalling and its impact on development and cancer. Nat Rev Cancer 2008, 8: 387] .
  • Second, tumor genome sequencing discovered the mutations in Wnt/B-catenin pathway components as well as epigenetic mechanisms that altered the expression of genes relevant to Wnt/ B-catenin pathway [Ying Y. et al. Epigenetic disruption of the WNT I beta-catenin signaling pathway in human cancers. Epigenetics 2009, 4:307] .
  • Wnt/B-catenin pathway also cooperates with other oncogenic signaling pathways in cancer and regulates tumorigenesis, growth, and metastasis [Klaus A, Birchmeier W. Wnt signalling and its impact on development and cancer. Nat Rev Cancer 8: 387-398, 2008].
  • WNT signaling between tumor and stromal cell interaction leading to tumorigenesis and metastasis [Shahi P, Park D, Pond AC, Seethammagari M, Chiou S-H, Cho K, et al. Activation of Wnt signaling by chemically induced dimerization of LRP5 disrupts cellular homeostasis.
  • PLoS ONE 2012, 7: e30814
  • stem- like colon cells with a high level of B-catenin signaling have a much greater tumorigenic potential than counterpart cells with low ⁇ -catenin signaling
  • ⁇ -catenin signaling Vermeulen L, De Sousa EMF, van der Heijden M, Cameron K, de Jong JH, Borovski T, Tuynman JB, Todaro M, Merz C, Rodermond H, Sprick MR, Kemper K, Richel DJ, Stassi G, Medema JP.
  • Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol. 2010, 12: 468].
  • activation of Wnt/6-catenin signalling pathway is also one of the major mechanism causing tumor recurrence and drug resistance. All these provide clear rationale to develop therapeutics targeting Wnt/6-catenin signaling pathway for the treatment of cancer.
  • Inhibition of TNKS blocks PARsylation of AXIN1 and AXIN2 and prevents their proteasomai degradation.
  • TNKS inhibition enhances the activity of the ⁇ -catenin destruction complex and suppresses 6- catenin nuclear transclocation and the expression of ⁇ -catenin target genes.
  • tankyrases are also implicated in other cellular functions, including telomere homeostasis, mitotic spindle formation, vesicle transport linked to glucose metabolism, and viral replication. In these processes, tankyrases interact with target proteins, catalyze poly (ADP-ribosyl)ation, and regulate protein interactions and stability.
  • TNKS1 controls telomere homeostasis, which promotes telomeric extension by PARsylating TRF1.
  • TRF1 is then targeted for proteasomai degradation by the E3 ubiquitin ligases F- box only protein 4 and/or RING finger LIM domain-binding protein (RLIM/RNF12), which facilitates telomere maintenance [Donigian JR and de Lange T. The role of the poly(ADP-ribose) polymerase tankyrase 1 in telomere length control by the TRF1 component of the shel terin complex. J Biol Chem 2007, 282:22662]. In addition, telomeric end-capping also requires canonical DNA repair proteins such as DNA-dependent protein kinase (DNAPK).
  • DNAPK DNA-dependent protein kinase
  • TNKS1 stabilizes the catalytic subunit of DNAPK (DNAPKcs) by PARsylation [Dregalla RC, Zhou J, Idate RR, Battaglia CL, Liber HL, Bailey SM. Regulatory roles of tankyrase 1 at telomeres and in DNA repair: suppression of T-SCE and stabilization of DNA-PKcs. Aging 2010, 2(10):691] . Altered expression of TNKS1 and/or TNKS2, as well as genetic alterations in the tankyrase locus, have been detected in multiple tumors, e.g.
  • tankyrases appear to have impact on viral infections. For example, in HSV infection, it was shown that the virus cannot replicate efficiently in cells with depletion of both TNKS1 and TNKS2 [Li I, Yamauchi Y,
  • TNKS1 knockout mice appeared to have reduced fat pads, suggesting a potential connection of TNKS and obesity. TNKS may also play a role in tissue fibrosis.
  • tankyrases are promising drug targets in regulating WNT signaling, telomere length (e.g. telomere shortening and DNA damage induced cell death), lung fibrogenesis, myelination and viral infection.
  • the invention presented here describes a novel class of tankyrase inhibitors and their potential clinical utility for the treatment of various diseases, such as cancer, aging, metabolic diseases (e.g. diabetes and obesity), fibrosis (e.g. lung fibrogenesis) and viral infection.
  • WO 2001 /000575 discloses heterocyclic dicarboxylic acid diamide derivatives as insecticides, including amido-substituted azole compounds.
  • R' represents -OR 9 , or -N(R ,0 )R n , which are as defined herein,
  • X 2 represents CR 6 or N, and R 4 , R 5 , R 6 , R 7 and R 8 are as defined herein, and
  • R 2 represents a group selected from hydrogen, d-Cralkyl, and CrCi-cycloalkyl ;
  • said compounds of the present invention have surprisingly been found to effectively inhibit TNKS1 and/or TNKS2 and may therefore be used for the treatment or prophylaxis of diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses or diseases which are accompanied with uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses mediated by TNKS1 and/or TNKS2 and/or mediated by the Wnt pathway, for example, haematological tumours, solid tumours, and/or metastases thereof, e.g.
  • leukaemias and myelodysplastic syndrome malignant lymphomas, head and neck tumours including brain tumours and brain metastases, tumours of the thorax including non-small cell and small cell lung tumours, gastrointestinal tumours, endocrine tumours, mammary and other gynaecological tumours, urological tumours including renal, bladder and prostate tumours, skin tumours, and sarcomas, and/or metastases thereof.
  • Compounds of the present invention may additionally show improved selectivity for TNKS1 and/or TNKS2 (e.g.
  • the present invention covers compounds of general formula (I) :
  • X 1 represents NR 3 or 0,
  • X 2 represents CR 6 or N
  • R 1 represents a group selected from :
  • R 2 represents a group selected from :
  • R 3 represents hydrogen
  • R 4 represents hydrogen
  • R 5 represents a group selected from :
  • R 6 represents a group selected from :
  • R 7 represents hydrogen
  • R 8 represents a group selected from :
  • aryl aryl-(Ci-C4-alkyl)-, heteroaryl, and heteroaryl- (Ci-Gralkyl)-,
  • aryl and heteroaryl groups are optionally substituted with one, two or three substituents, which are independently of each other selected from : G-G-alky , G-Gralkoxy, G-Grhydroxyalkyl, GrG-cycloalkyl, G-G-cycloalkoxy, G-Grhaloalkyl, G-G-haloalkoxy, halogen, cyano, nitro, hydroxy, -N(R'°)R" , R'°(R" )N-(G-G.-alkyl)-, R 10 (R 11 )N-(C 2 -C 4 -alkoxy)-,
  • R 9 represents G-Gralkyl
  • R 10 and R" are independently of each other selected from :
  • G-G-hydroxyalkyl (G-G-alkoxy)-(G-G-alkyl)-, G-G-haloalkyl, H 2 N-(GrG-alkyl)- , (G -G- alkyl)N(H)(G-G-alkyl)-, (G-G-alkyl) 2 N(G-G-alkyl)-, 4-6 membered heterocycloalkyl, (4-6 membered heterocycloalkyl)- (G-G-alkyl)-,
  • 4-6-membered heterocycloalkyl groups are optionally substituted with one or two substituents, which are independently of each other selected from :
  • R 12 represents a group selected from :
  • each definition is independent.
  • R 10 , R 11 , and/or R 12 occur more than one time in any compound of formula (I ) each definition of R'°, R" , and R 12 is independent.
  • a hyphen at the beginning or at the end of the constituent marks the point of attachment to the rest of the molecule. Should a ring be substituted the substitutent could be at any suitable position of the ring, also on a ring nitrogen atom if suitable.
  • halogen halogen atom
  • halo- halo- or Hal-
  • fluorine chlorine, bromine or iodine atom.
  • Ci-Ct-alkyl is to be understood as preferably meaning a linear or branched, saturated, monovalent hydrocarbon group having 1 , 2, 3, or 4, carbon atoms, e.g. a methyl, ethyl, propyl, butyl, iso-propyl, iso-butyl, sec-butyl, tert-butyl group, more particularly 1 , 2 or 3 carbon atoms ("G -Cralkyl ”) , e.g. a methyl, ethyl, n-propyl- or iso- propyl group, even more particularly 1 or 2 carbon atoms ("Ci -Cj-alkyl”), e.g. a methyl, ethyl group.
  • C2-C4-alkyl is to be understood as preferably meaning a linear or branched , saturated, monovalent hydrocarbon group having 2, 3 , or 4, carbon atoms, e.g. a ethyl, propyl, butyl, iso-propyl, iso-butyl, sec-butyl, tert-butyl group, more particularly 2 or 3 carbon atoms e.g. a ethyl, n-propyl- or iso-propyl group, even more particularly 2 carbon atoms (“Cralkyl”) , i. e. a ethyl group.
  • G-Cs-hydroxya kyl is to be understood as preferably meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "G -Cralkyl” is defined supra, and in which one or more hydrogen atoms is replaced by a hydroxy group, e.g. a hydroxymethyl, 1 -hydroxyethyl, 2-hydroxyethyl, 1 ,2-dihydroxyethyl, 3- hydroxypropyl , 2-hydroxypropyl, 2, 3-dihydroxypropyl, 1 , 3-dihydroxypropan-2-yl group.
  • G-G-hydroxyalkyl is to be understood as preferably meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "G-G-alkyl” is defined supra, and in which one or more hydrogen atoms is replaced by a hydroxy group, e.g. a 2-hydroxyethyl, 3-hydroxypropyl, 2-hydroxypropyl, 2, 3-dihydroxypropyl, 3-hydroxy- 2-methyl-propyl , 2-hydroxy-2-methyl-propyl group.
  • CrC4-haloalkyl is to be understood as preferably meaning a linear or branched , saturated, monovalent hydrocarbon group in which the term "G-G-alkyl” is defined supra, and in which one or more hydrogen atoms is replaced by a halogen atom , in identically or differently, i. e. one halogen atom being independent from another. Particularly, said halogen atom is F.
  • Said G-G-ha oa kyl group is, for example, CF), -CHF2, -CH2F, -CF2CF3, - CH2CH2F, -CH2CHF2, -CH2CF3, or -CH2CH2CF3.
  • G -G-alkoxy is to be understood as preferably meaning a linear or branched, saturated , monovalent, hydrocarbon group of formula 0-alkyl having 1 , 2 , or 3 carbon atoms, in which the term “alkyl” is defined supra, e.g. a methoxy, ethoxy, n-propoxy, or iso-propoxy group, or an isomer thereof.
  • G-Crhaloalkoxy is to be understood as preferably meaning a linear or branched, saturated, monovalent G-Ci-alkoxy group, as defined supra, in which one or more of the hydrogen atoms is replaced, in identically or differently, by a halogen atom. Particularly, said halogen atom is F.
  • Said G-Crhaloalkoxy group is, for example, OCF3, - OCHF?, -OCH7F, -OCF 2 CF 3) or -OCH2CF3.
  • C3-C -cycloalkyl is to be understood as meaning a saturated, monovalent, monocyclic hydrocarbon ring which contains 3, or 4, carbon atoms (“CrG-cycloalkyl”).
  • Said G-G-cyc oa ky group is for example, a monocyclic hydrocarbon ring, e.g. a cyclopropyl, or cyclobutyl ring.
  • C3-G-cycloalkoxy is to be understood as preferably meaning a saturated, monovalent, hydrocarbon ring which contains 3. or 4 carbon atoms of formula 0- cycloalkyl, in which the term "cycloalkyl” is defined supra, e.g. a cyclopropyloxy, or cyclobutyloxy.
  • said heterocycloalkyl can be a 4-membered ring, such as an azetidinyl, oxetanyl, or a 5-membered ring, such as tetrahydrofuranyl, dioxolinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, or a 6-membered ring, such as tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, or N- methylpiperazinyl.
  • said heterocycloalkyl can be benzo fused.
  • 4- to 6-membered heterocycloalkyl can be selected from piperazinyl, tetrahydro-2H-pyranyl, tetrahydrofuranyl, pyrrolidinyl, piperidinyl, morpholinyl, azetidinyl, 2-oxoimidazolidinyl, 2-oxopyrrolidinyl and 1 , 1 - dioxidothiomorpholinyl.
  • 4- to 6- membered heterocycloalkyl can be selected from piperazin-1 -yl, tetrahydro-2H-pyran-4-yl, tetrahydrofuran-3-yl, pyrrolidin-1 -yl, pyrrolidin-2-yl, pyrrolidin-3-yl, piperidin-4-yl, piperidin-1 -yl, piperidin-2-yl, piperidin-3-yl, morpholin-4-yl, azetidin-1 -yl, tetrahydrofuran-2-yl, 2-oxoimidazolidin-1 -yl, 2-oxopyrrolidin-1 -yl and 1 , 1 - dioxidothiomorpholin-4-yl.
  • aryl is to be understood as preferably meaning a monovalent, aromatic or partially aromatic, mono- or bicyclic hydrocarbon ring having 6, 7, 8, 9 or 10 carbon atoms (a "C6-Cio-aryl” group), particularly a ring having 6 carbon atoms (a "Ce-aryl” group), e.g. a phenyl group; or a ring having 9 carbon atoms (a "Cg-aryl” group), e.g. an indanyl or indenyl group, or a ring having 10 carbon atoms (a "Cio-aryl” group), e.g. a tetralinyl, dihydronaphthyl, or naphthyl group.
  • aryl is phenyl.
  • heteroaryl is understood as preferably meaning a monovalent, monocyclic aromatic ring system having 5 or 6 ring atoms (a "5- to 6-membered heteroaryl” group), which contains at least one heteroatom which may be identical or different, said heteroatom being such as oxygen, nitrogen, NH or sulfur.
  • heteroaryl is selected from thienyl, furanyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl etc.
  • heteroaryl can be selected from pyrazolyl, thienyl, pyridyl, furanyl, thiazolyl, oxazolyl, and pyrazinyl.
  • the heteroarylic or heteroarylenic radicals include all the possible isomeric forms thereof, e.g. the positional isomers thereof.
  • the term pyridinyl or pyridinylene includes pyridin-2-yl, pyridin-2-ylene, pyridin-3-yl, pyridin-3-ylene, pyridin-4-yl and pyridin-4-ylene; or the term thienyl or thienylene includes thien-2-yl, thien-2-ylene, thien-3-yl and thien-3- ylene.
  • heteroarylic radicals include all the possible isomeric forms thereof, e.g. the positional isomers thereof.
  • pyridinyl includes pyridin-2-yl, pyridin-3-yl and pyridin-4-yl.
  • aromatic and non-aromatic (hetero)cyclic groups may optionally be substituted as defined herein.
  • the substituents may be present both when said aromatic and non-aromatic (hetero)cyclic groups exist as a (unitary) constituent, such as, for example, G-G-cycloalkyl, 4- to 6-membered heterocycloalkyl, aryl and heteroaryl groups, or as part of a constituent composed of more than one part, such as, for example, (G-G-cycloalkyl)-G-G-alkyl-, (4- to 6-membered heterocycloalkyl)- (CrC -alkyl)- , aryl-(G-G-alkyl)-, and heteroaryl- (C i -G-alkyl )- , for example.
  • the present invention includes all suitably substituted aromatic and non-aromatic (hetero)cyclic groups both as a (unitary) constituent, or as part of a constituent composed of more than one part.
  • aromatic and non-aromatic (hetero)cyclic groups both as a (unitary) constituent, or as part of a constituent composed of more than one part.
  • suitable is to be understood as meaning chemically possible to be made by methods within the knowledge of a skilled person.
  • Ci-G as used throughout this text, e.g. in the context of the definition of "G- G-alkyl", or “G-G-haloalkyl”, is to be understood as meaning an alkyl group having a finite number of carbon atoms of 1 to 4, i.e. 1 , 2, 3, or 4 carbon atoms. It is to be understood further that said term “G-G,” is to be interpreted as any sub-range comprised therein, e.g. G-G. , G- , G-G , G-G , G-G ; particularly G-G , G-G , G-G. Similarly, as used herein, the term "C2-C4", as used throughout this text, e.g.
  • C2-C4-alkyl in the context of the definitions of "C2-C4-alkyl", and "C2-C4-hydroxyalkyl” is to be understood as meaning an a Iky I group or a hydroxyalkyl group having a finite number of carbon atoms of 2 to 4, i. e. 2, 3, or 4 carbon atoms. It is to be understood further that said term "C2-C4" is to be interpreted as any sub-range comprised therein, e.g. C2-C4 , C3-C4 , C2-C3; particularly
  • substituted means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded , and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • Ring system substituent means a substituent attached to an aromatic or nonaromatic ring system which, for example, replaces an available hydrogen on the ring system.
  • the term "one or more”, e.g. in the definition of the substituents of the compounds of the general formulae of the present invention, is understood as meaning “one, two, three, four or five, particularly one, two, three or four, more particularly one, two or three, even more particularly one or two".
  • the invention also includes all suitable isotopic variations of a compound of the invention.
  • An isotopic variation of a compound of the invention is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually or predominantly found in nature.
  • isotopes that can be incorporated into a compound of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine, chlorine, bromine and iodine, such as H (deuterium), 3 H (tritium), "C, 13 C, 14 C, 15 N, ,7 0, 18 0, 32 P, 33 P, 3 S, 34 S, 35 S, 36 S, 18 F, 6Cl, 82 Br, 123 l, 124 l, 125 l, 129 l and 131 l, respectively.
  • isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine, chlorine, bromine and iodine, such as H (deuterium), 3 H (tritium), "C, 13 C, 14 C, 15 N, ,7 0, 18 0, 32 P, 33 P, 3 S, 34 S, 35 S, 36 S, 18 F, 6Cl, 82 Br, 123 l, 124
  • isotopic variations of a compound of the invention are useful in drug and/or substrate tissue distribution studies. Tritiated and carbon-14, i.e. , 14 C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence is preferred in some circumstances.
  • isotopic variations of a compound of the invention can generally be prepared by conventional procedures known by a person skilled in the art such as by the illustrative methods or by the preparations described in the examples hereafter using appropriate isotopic variations of suitable reagents.
  • stable compound' or “stable structure” is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • the compounds of this invention optionally contain one or more asymmetric centre, depending upon the location and nature of the various substituents desired.
  • Asymmetric carbon atoms is present in the (R) or (S) configuration , resulting in racemic mixtures in the case of a single asymmetric centre, and diastereomeric mixtures in the case of multiple asymmetric centres.
  • asymmetry may also be present due to restricted rotation about a given bond , for example, the central bond adjoining two substituted aromatic rings of the specified compounds.
  • the compounds of the present invention optionally contain sulphur atoms which are asymmetric, such as an asymmetric sulfoxide, of structure: , for example,
  • Preferred compounds are those which produce the more desirable biological activity.
  • Separated, pure or partially purified isomers and stereoisomers or racemic or diastereomeric mixtures of the compounds of this invention are also included within the scope of the present invention.
  • the purification and the separation of such materials can be accomplished by standard techniques known in the art.
  • the optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example, by the formation of diastereoisomeric salts using an optically active acid or base or formation of covalent diastereomers.
  • appropriate acids are tartaric, diacetyl tartaric, ditoluoyltartaric and camphorsulfonic acid.
  • Mixtures of diastereoisomers can be separated into their individual diastereomers on the basis of their physical and/or chemical differences by methods known in the art, for example, by chromatography or fractional crystallisation.
  • the optically active bases or acids are then liberated from the separated diastereomeric salts.
  • a different process for separation of optical isomers involves the use of chiral chromatography (e.g. , chiral HPLC columns), with or without conventional derivatisation, optimally chosen to maximise the separation of the enantiomers.
  • Suitable chiral HPLC columns are manufactured by Daicel, e.g. , Chiracel OD and Chiracel OJ among many others, all routinely selectable.
  • Enzymatic separations, with or without derivatisation are also useful.
  • the optically active compounds of this invention can likewise be obtained by chiral syntheses utilizing optically active starting materials.
  • the present invention includes all possible stereoisomers of the compounds of the present invention as single stereoisomers, or as any mixture of said stereoisomers, e.g. R- or S- isomers, or E- or Z-isomers, in any ratio.
  • Isolation of a single stereoisomer, e.g. a single enantiomer or a single diastereomer, of a compound of the present invention is achieved by any suitable state of the art method , such as chromatography, especially chiral chromatography, for example.
  • the compounds of the present invention may exist as tautomers.
  • any compound of the present invention which contains a pyrazole moiety as a heteroaryl group for example can exist as a 1 H tautomer, or a 2H tautomer, or even a mixture in any amount of the two tautomers, namely :
  • the present invention can exist as one of the below tautomers, or even in a mixture in any amount of the two tautomers, namely:
  • the present invention includes all possible tautomers of the compounds of the present invention as single tautomers, or as any mixture of said tautomers, in any ratio.
  • the compounds of the present invention can exist as N-oxides, which are defined in that at least one nitrogen of the compounds of the present invention is oxidised.
  • the present invention includes all such possible N-oxides.
  • the present invention also relates to useful forms of the compounds as disclosed herein, such as metabolites, hydrates, solvates, prodrugs, salts, in particular pharmaceutically acceptable salts, and co-precipitates.
  • the compounds of the present invention can exist as a hydrate, or as a solvate, wherein the compounds of the present invention contain polar solvents, in particular water, methanol or ethanol for example as structural element of the crystal lattice of the compounds.
  • polar solvents in particular water, methanol or ethanol for example as structural element of the crystal lattice of the compounds.
  • the amount of polar solvents, in particular water may exist in a stoichiometric or non-stoichiometric ratio.
  • stoichiometric solvates e.g. a hydrate, hemi-, (semi-), mono- , sesqui-, di- , tri- , tetra- , penta- etc. solvates or hydrates, respectively, are possible.
  • the present invention includes all such hydrates or solvates.
  • the compounds of the present invention can exist in free form, e.g. as a free base, or as a free acid, or as a zwitterion, or can exist in the form of a salt.
  • Said salt may be any salt, either an organic or inorganic addition salt, particularly any pharmaceutically acceptable organic or inorganic addition salt, customarily used in pharmacy.
  • pharmaceutically acceptable salt refers to a relatively non-toxic, inorganic or organic acid addition salt of a compound of the present invention.
  • pharmaceutically acceptable salt refers to a relatively non-toxic, inorganic or organic acid addition salt of a compound of the present invention.
  • SCO S. M. Berge, er al. “Pharmaceutical Salts, " J. Pharm. Sci. 1977, 66, 1 -19.
  • a suitable pharmaceutically acceptable salt of the compounds of the present invention may be, for example, an acid-addition salt of a compound of the present invention bearing a nitrogen atom, in a chain or in a ring, for example, which is sufficiently basic, such as an acid-addition salt with an inorganic acid, such as hydrochloric, hydrobromic, hydroiodic, sulfuric, bi sulfuric, phosphoric, or nitric acid, for example, or with an organic acid, such as formic, acetic, acetoacetic, pyruvic, trifluoroacetic, propionic, butyric, hexanoic, heptanoic, undecanoic, lauric, benzoic, salicylic, 2-(4-hydroxybenzoyl)-benzoic, camphoric, cinnamic, cyclopentanepropionic, digluconic, 3-hydroxy-2-naphthoic, nicotinic, pamoic, pectinic,
  • an alkali metal salt for example a sodium or potassium salt
  • an alkaline earth metal salt for example a calcium or magnesium salt
  • an ammonium salt or a salt with an organic base which affords a physiologically acceptable cation, for example a salt with N-methyl-glucamine, dimethyl-glucamine, ethyl-glucamine, lysine, dicyclohexylamine, 1 ,6-hexadiamine, ethanolamine, glucosamine, sarcosine, serinol, tris-hydroxy-methyl-aminomethane, aminopropandiol, sovak-base, 1 -amino-2, 3,4- butantriol.
  • basic nitrogen containing groups may be quaternised with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides ; dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate ; and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and strearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate
  • diamyl sulfates long chain halides such as decyl, la
  • acid addition salts of the claimed compounds may be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods.
  • alkali and alkaline earth metal salts of acidic compounds of the invention are prepared by reacting the compounds of the invention with the appropriate base via a variety of known methods.
  • the present invention includes all possible salts of the compounds of the present invention as single salts, or as any mixture of said salts, in any ratio.
  • in vivo hydrolysable ester is understood as meaning an in vivo hydrolysable ester of a compound of the present invention containing a carboxy or hydroxy group, for example, a pharmaceutically acceptable ester which is hydrolysed in the human or animal body to produce the parent acid or alcohol.
  • suitable pharmaceutically acceptable esters for carboxy include for example alkyl, cycloalkyl and optionally substituted phenylalkyl, in particular benzyl esters, G-Ce alkoxymethyl esters, e.g. methoxymethyl, G-Ce alkanoyloxymethyl esters, e.g.
  • An in vivo hydrolysable ester of a compound of the present invention containing a hydroxy group includes inorganic esters such as phosphate esters and [alpha] -acyloxyalkyl ethers and related compounds which as a result of the in vivo hydrolysis of the ester breakdown to give the parent hydroxy group.
  • inorganic esters such as phosphate esters and [alpha] -acyloxyalkyl ethers and related compounds which as a result of the in vivo hydrolysis of the ester breakdown to give the parent hydroxy group.
  • [alpha] -acyloxyalkyl ethers include acetoxymethoxy and 2,2-dimethylpropionyloxymethoxy.
  • a selection of in vivo hydrolysable ester forming groups for hydroxy include alkanoyl, benzoyl, phenylacetyl and substituted benzoyl and phenylacetyl, alkoxycarbonyl (to give alkyl carbonate esters), dialkylcarbamoyl and N-(dialkylaminoethyl)-N-alkylcarbamoyl (to give carbamates), dialkylaminoacetyl and carboxyacetyl.
  • the present invention covers all such esters.
  • the present invention includes all possible crystalline forms, or polymorphs, of the compounds of the present invention, either as single polymorph, or as a mixture of more than one polymorph, in any ratio.
  • the present invention covers compounds of general formula (I), supra, in which :
  • X 1 represents NR ;t ,
  • X 2 represents CR 6 ,
  • R' represents a group selected from :
  • R 2 represents a group selected from :
  • R 3 represents hydrogen
  • R 4 represents hydrogen
  • R 5 represents a group selected from :
  • R 6 represents a group selected from :
  • R 7 represents hydrogen
  • R 8 represents a group selected from :
  • aryl aryl-(CrC4-alkyl)-, heteroaryl, and heteroaryl- (Ci -C4-alkyl)-,
  • aryl and heteroaryl groups are optionally substituted with one, two or three substituents, which are independently of each other selected from :
  • G-G-alkyl G-Cs-a koxy, G-Cs-hydroxya kyl, G-Grhaloalkyl, halogen, cyano, -N(R 10 )R 11 ,
  • R 9 represents G-G-alkyl
  • R'° and R" are independently of each other selected from :
  • G-Grhydroxyalkyl (Ci-alkoxy)- (C2-C3-alkyl)-, G-G-ha oalky , HjN-iGi-G-alkyl)-, (G- alkyl)N(H)(G-G-alkyl)-, (G-alkyl) 2 N(G-G-alkyl)-, (4-6 membered heterocycloalkyl)- (G-G- alkyl)-, wherein 4-6-membered heterocycloalkyl groups are optionally substituted with one or two substituents, which are independently of each other selected from :
  • R' 2 represents a group selected from :
  • the present invention covers compounds of general formula (I ), supra, in which :
  • X 1 represents NR 3 ,
  • X 2 represents CR 6 ,
  • R 1 represents -N (R , 0 )R" ,
  • R 2 represents hydrogen
  • R 3 represents hydrogen
  • R 4 represents hydrogen
  • R 5 represents hydrogen
  • R0 represents hydrogen
  • R 7 represents hydrogen
  • R 8 represents a group selected from
  • aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
  • R'° and R 11 are independently of each other selected from :
  • Ci -G-alkyl C3-C4-cycloalkyl, (C3-C4-cycloalkyl)-(Ci -alkyl)-,
  • the present invention covers compounds of general formula (I ), supra, in which :
  • X 1 represents NR 3
  • X 2 represents CR 6
  • R' represents -N(R 10 )R 11 ,
  • R 2 represents hydrogen
  • R 3 represents hydrogen
  • R4 represents hydrogen
  • R 5 represents hydrogen
  • R 6 represents hydrogen
  • R 7 represents hydrogen
  • R 8 represents a group selected from
  • aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
  • R 10 and R" are independently of each other selected from :
  • the present invention covers a compound of general formula (I), supra, which is selected from the group consisting of :
  • the invention relates to compounds of formula (I), wherein :
  • X 1 represents NR 3 ,
  • X 2 represents CR 6 ,
  • R 1 represents -N(R !0 )R
  • R 2 represents hydrogen
  • R 3 represents hydrogen
  • R 4 represents hydrogen
  • R 5 represents hydrogen
  • R 6 represents hydrogen
  • R 7 represents hydrogen
  • R 8 represents a group selected from
  • aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
  • R'° and R 11 are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • X 1 represents NR ⁇
  • X 2 represents CR 6 ,
  • R 1 represents -N (R , 0 )R" ,
  • R 2 represents hydrogen
  • R 3 represents hydrogen
  • R 4 represents hydrogen
  • R 5 represents hydrogen
  • R 6 represents hydrogen
  • R 7 represents hydrogen
  • R 8 represents a group selected from
  • aryl is optionally substituted with one, or two substituents, which are independently of each other selected from :
  • R'° and R" are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • X 1 represents NR 3 or 0.
  • the invention relates to compounds of formula (I), wherein :
  • X 1 represents NR 3 .
  • the invention relates to compounds of formula (I), wherein :
  • X 1 represents 0.
  • the invention relates to compounds of formula (I), wherein :
  • X 2 represents CR 6 or N ,
  • the invention relates to compounds of formula (I), wherein :
  • X 2 represents CR 6 .
  • the invention relates to compounds of formula (I), wherein :
  • X 2 represents N.
  • the invention relates to compounds of formula (I), wherein :
  • R 1 represents a group selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 1 represents a group selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 1 represents a group selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 2 represents a group selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 2 represents a group selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 2 represents hydrogen
  • the invention relates to compounds of formula (I), wherein :
  • R 5 represents a group selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 5 represents a group selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 5 represents hydrogen
  • the invention relates to compounds of formula (I), wherein :
  • R 6 represents a group selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 6 represents a group selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 6 represents hydrogen.
  • the invention relates to compounds of formula (I), wherein :
  • R 8 represents a group selected from :
  • aryl and heteroaryl groups are optionally substituted with one, two or three substituents, which are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 8 represents a group selected from :
  • aryl aryl-(Ci-Ct-alkyl)-, heteroaryl, and heteroaryl- (G-Oalkyl)-,
  • aryl and heteroaryl groups are optionally substituted with one, two or three substituents, which are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 8 represents a group selected from :
  • aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 8 represents a group selected from :
  • aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 8 represents aryl optionally substituted with one, two or three substituents, which are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 8 represents aryl-(Ci -Cralkyl)- wherein aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 8 represents a group selected from :
  • aryl is optionally substituted with one, or two substituents, which are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 8 aryl, wherein aryl is optionally substituted with one, or two substituents, which are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 8 represents aryl-(Ci -G-alkyl)-, wherein aryl is optionally substituted with one, or two substituents, which are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 9 represents Ci-Cj-alkyl.
  • the invention relates to compounds of formula (I), wherein :
  • R 9 represents Ci -C 2 -alkyl.
  • the invention relates to compounds of formula (I), wherein :
  • R'° and R 11 are independently of each other selected from :
  • Ci Ci -Ca-alkyl, C3-C4-cycloalkyl, (C3-C4-cycloalkyl)-(0-C4-alkyl)-,
  • Ci-alkyl O-haloalkyl, Ci -alkoxy, Ci -haloalkoxy, halogen, and hydroxyl.
  • the invention relates to compounds of formula (I), wherein :
  • R'° and R 11 are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R'° is selected from hydrogen, and G -Ct-alkyl, preferably hydrogen,
  • R" is selected from :
  • 4- to 6-membered heterocycloalkyl groups are optionally substituted with one or two substituents, which are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • said 4-to 6-membered heterocycloalkyl group being optionally substituted with one or two substituents, which are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R'° and R" are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R'° is selected from hydrogen, and Ci -Ca-alkyl, preferably hydrogen,
  • R" is selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R'° and R 11 are independently of each other selected from : hydrogen, Ci-Cj-alkyl, CrC4-cycloalkyl, (CrC4-cycloalkyl)-(G-alkyl)-,
  • the invention relates to compounds of formula (I), wherein :
  • R 10 is selected from hydrogen, and G-G-alkyl, preferably hydrogen,
  • R" is selected from :
  • G-G-hydroxyalkyl (G-alkoxy)- (G-G-alkyl)-, G-Grhaloalkyl, H2N-(Cralkyl)-, (G- alkyl)N(H)(G-alkyl)-, (Ci-alkyl) 2 N(C2-alkyl)-, (6-membered heterocycloalkyl)-(C 2 -alkyl)-.
  • the invention relates to compounds of formula (I), wherein :
  • R 10 and R" are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R'° is selected from hydrogen, and CrC 2 -alkyl, preferably hydrogen,
  • R 11 is selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R'° and R" are independently of each other selected from :
  • R'° and R" are independently of each other selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 10 is selected from hydrogen, and Ci-Cralkyl, preferably hydrogen,
  • R n is selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R'° is selected from hydrogen, and Ci-C?-alkyl, preferably hydrogen,
  • R" is selected from :
  • R' 2 represents a group selected from :
  • the invention relates to compounds of formula (I), wherein :
  • R 12 represents a group selected from :
  • R 12 represents a group selected from :
  • the invention relates to compounds of formula (I), according to any of the above-mentioned embodiments, in the form of or a stereoisomer, a tautomer, an N- oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.
  • the present invention covers compounds of general formula (I) which are disclosed in the Example section of this text, infra.
  • the present invention covers methods of preparing compounds of the present invention, said methods comprising the steps as described in the Experimental Section herein.
  • another aspect of the present invention relates to a method of preparing a compound of general formula (I) as defined herein, said method comprising the step of allowing an intermediate compound of general formula (III) :
  • Another aspect of the present invention relates to a method of preparing a compound of general formula (I) as defined herein, said method comprising the step of allowing an intermediate compound of general formula (3-4) :
  • Another aspect of the present invention relates to a method of preparing a compound of general formula (I ) as defined, said method comprising the step of allowing an intermediate compound of general formula (VI) : in which Xi represents NR 3 , X 2 , R 2 , R 3 , R 4 , R 5 , R 7 and R 8 are as defined herein for the compound of general formula (I),
  • Another aspect of the present invention relates to a method of preparing a compound of general formula (I) as defined herein, said method comprising the step of allowing an intermediate compound of general formula (IV) : in which X 1 represents N , and X 2 , R 2 , R 4 , R 5 , R 7 and R 8 are as defined herein for the compound of general formula (I),
  • X 1 represents NR 3 and X 2 , R ⁇ R 2 , R 3 , R 4 , R 5 , R 7 , and R 8 are as defined herein for the compound of general formula (I ).
  • Another aspect of the present invention relates to a method of preparing a compound of general formula (I ) as defined herein , said method comprising the step of allowing an intermediate compound of general formula (VII ) : in which X 2 , R 4 , R 5 , R 7 , and R 8 are as defined herein for the compound of general formula (I), to react with a compound of general formula (4-1 ) :
  • Another aspect of the invention is intermediate (3-4) or a salt thereof:
  • Xt represents NR 3 , X 2 , R ⁇ R 2 , R 3 , R 4 , R 5 , and R 9 are as defined herein for the compound of general formula (! ) and R 13 represents R 9 , or H.
  • Another aspect of the invention is intermediate (VI ) or a salt thereof:
  • Another aspect of the invention is intermediate (VII) or a salt thereof:
  • the present invention covers the use of the intermediate compounds of general formula (III), (3-4), (VI), (IV) and (VII), or a salt thereof for the preparation of a compound of general formula (I)
  • Another aspect of the invention relates to the intermediates or salts thereof described herein and their use for preparing a compound of formula (I) as defined supra or an N- oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
  • Aromatic amines as intermediates for the synthesis of compounds of the invention are either commercially available or can be synthesized as depicted in scheme 1 .
  • Scheme 1 Synthesis of aromatic amines, wherein X 2 , R 4 , R 5 , R 7 and R 8 are as defined for the compound of general formula (I) supra, and in which PG represents a protecting group, such as a BOC group, and W represents a hydroxy group or a chlorine atom.
  • PG represents a protecting group, such as a BOC group
  • W represents a hydroxy group or a chlorine atom.
  • amides of type 1 -3 can be obtained by reaction with amines of type 1-2 in the presence of a coupling agent such as, for example, HATU or PyBOP, or via the formation of the corresponding acid chloride of type 1 - 1.
  • a coupling agent such as, for example, HATU or PyBOP
  • amides of type 1 -3 can be obtained by reaction with amines of type 1 -2 in the presence of a coupling agent such as, for example, HATU or PyBOP, or via the formation of the corresponding acid chloride of type 1 - 1.
  • a coupling agent such as, for example, HATU or PyBOP
  • Scheme 2 Synthesis of 3,4 dicarboxylic acid substituted imidazoles, wherein X 1 represents NR 3 , and R 2 and R 3 are as defined for the compounds of general formula (I) supra.
  • benzene-1 ,2-diamine 2-1 can be reacted with carboxyclic acids at elevated temperatures to give compounds of type 2-2.
  • Scheme 3 Synthesis of compounds starting from dicarboxylic acid precursors, wherein Xt represents NR ⁇ except for compounds 3-1 and (III) wherein X 1 represents N, and X 2 , R' , R 2 , R ⁇ R 4 , R 5 , R 7 , R 8 , R 9 , R'° and R 1 1 are as defined for the compound of general formula (I) supra, and R 13 represents R 9 or H.
  • Compounds of general formula (I) can be obtained directly by reacting compounds of general formula (III) with a fully decorated aromatic amine of general formula (II) at room temperature or at elevated temperature (e.g. under reflux).
  • an intermediate of type 3-4 can be obtained by reacting a compound of general formula (III) with a suitably substituted aromatic amine of type 3-3 at room temperature or elevated temperature (e.g. under reflux).
  • compounds of general formula (I) can be obtained in a 2 step procedure from esters of type 3-4a via, first, ester hydrolysis to afford carboxylic acid 3-4b, for example, under basic conditions, followed by standard amide bond forming reactions, for example with amines of type 1 -2 in the presence of a coupling agent such as, for example, T3P, HATU, PyBOP, or alternatively in a three step procedure after hydrolysis of the ester, generation of corresponding acid chloride, for example using thionylchloride or 1 -chloro- N, N,2-tri methyl- 1 -propenylamine and reaction with amines of type 1 -2 under basic conditions in presence of , for example, N, N-diisopropylethylamine or pyridine.
  • a coupling agent such as, for example, T3P, HATU, PyBOP
  • a base such as, for example, N,N-diisopropylethylamine
  • Compounds of general formula (IV) can be transformed to compounds of general formula (I) by reaction with nucleophiles of type 3-2, such as, for example, amines HN(R'°)R" , or alcohols HOR 9 , optionally in the presence of a base, such as, for example N, N- diisopropylethylamine.
  • nucleophiles of type 3-2 such as, for example, amines HN(R'°)R" , or alcohols HOR 9 , optionally in the presence of a base, such as, for example N, N- diisopropylethylamine.
  • Compounds of general formula (VI) can be transformed into amides of general formula (I), according to the invention, for example by treatment with different amines of type HN(R ,0 )(R , , ) ) optionally in presence of a base, such as, for example, N,N- diisopropylethylamine.
  • a base such as, for example, N,N- diisopropylethylamine.
  • R 1 N(R 10 )R 11
  • a base such as, for example, N,N- diisopropylethylamine
  • a base such as, for example,N- diisopropy
  • Chemical names were generated using ACD/Name Batch Version 12.02.. In case there is discrepancy between the chemical name of a compound and its chemical structure, the chemical structure shall prevail. In some cases generally accepted names of commercially available reagents were used in place of ACD generated names.
  • NMR peak forms are stated as they appear in the spectra, possible higher order effects have not been considered.
  • the 1 H-NMR data of selected examples are listed in the form of ⁇ -NMR peaklists. For each signal peak the ⁇ value in ppm is given, followed by the signal intensity, reported in round brackets. The ⁇ value-signal intensity pairs from different peaks are separated by commas. Therefore, a peaklist is described by the general form: ⁇ (intensity! ), ⁇ ? (intensity?), ... , ⁇ ,- (intensity.), ... , ⁇ ⁇ (intensity,,).
  • the intensity of a sharp signal correlates with the height (in cm) of the signal in a printed NMR spectrum. When compared with other signals, this data can be correlated to the real ratios of the signal intensities. In the case of broad signals, more than one peak, or the center of the signal along with their relative intensity, compared to the most intense signal displayed in the spectrum, are shown.
  • a ⁇ -NMR peaklist is similar to a classical ' H-NMR readout, and thus usually contains all the peaks listed in a classical NMR interpretation.
  • peaklists can show solvent signals, signals derived from stereoisomers of target compounds (also the subject of the invention), and/or peaks of impurities.
  • the peaks of stereoisomers, and/or peaks of impurities are typically displayed with a lower intensity compared to the peaks of the target compounds (e.g. , with a purity of >90%).
  • Such stereoisomers and/or impurities may be typical for the particular manufacturing process, and therefore their peaks may help to identify the reproduction of our manufacturing process on the basis of "by-product fingerprints".
  • An expert who calculates the peaks of the target compounds by known methods can isolate the peaks of target compounds as required , optionally using additional intensity filters. Such an operation would be similar to peak-picking in classical ⁇ -NMR interpretation.
  • Method 2lnstrument Waters Acquity UPLCMS SingleQuad; Column: Acquity UPLC BEH C18 1 .7 ⁇ , 50x2.1 mm; eluent A: water + 0.2 vol % aqueous ammonia (32%), eluent B: acetonitrile; gradient: 0-1 .6 min 1 -99% B, 1 .6-2.0 min 99% B; flow 0.8 ml/ min; temperature: 60 C; DAD scan: 210-400 nm.
  • Method 4 Instrument: Waters Acquity UPLCMS Tof; column: Kinetex C 18 (Phenomenex) 2.6 ⁇ , 50x2.1 mm; eluent A: water + 0.05 Vol-% formic acid (99%), eluent B: acetonitrile + 0.05% formic acid; gradient: 0-0.2 min 98% A, 0.2-1 .7 min 98-10% A, 1 .7-1.9 min 10% A, 1.9-2.0 min 10-98% A, 2.0-2.5 min 98% A; flow 1.3 ml/min; temperature: 60 C; DAD scan: 210-400 nm
  • Instrument Waters Autopurification MS SingleQuad; Column: Waters XBrigde C18 5 ⁇ 100x30mm; eluent A: water + 0.1 vol % formic acid (99%), eluent B: acetonitrile; gradient eluent A/ eluent B, flow 70 ml/min; temperature: 25 C; DAD scan: 210-400 nm.
  • Benzotriazol-1 -yl-oxytripyrrolidinophosphonium hexafluorophosphate (1 17 mg, 225 mol) and N.N-diisopropylethylamine (140 ⁇ , 820 mol) were added to a mixture of lithium 4- ⁇ [(4- ⁇ [2-(piperidin-1-yl)ethy[]carbamoy[ ⁇ -1H-imidazo[-5-yl)carbony[]amino ⁇ benzoate (100 mg, 80 % purity, 204 ⁇ ) and 1 -(3-chlorophenyl)methanamine (31.8 mg, 225 ⁇ ) and the mixture was stirred at room temperature for 12 h. For work-up, the reaction mixture was concentrated and the residue was purified by preparative HPLC (Method 6) followed by recrystallization from ethyl acetate to give the title compound (12.5 mg).
  • reaction mixture was concentrated and the residue was stirred with ethanol.
  • reaction mixture was concentrated and the residue was purified by flash column chromatography (25 g Snap cartridge, dichloromethane/ methanol-gradient, 20% -> 100% methanol) followed by trituration with dichloromethane to give the title compound (130 mg, 41%yield).
  • reaction mixture was concentrated and the residue was purified by flash column chromatography (25 g Snap cartridge, dichloromethane/ methanol-gradient, 20% -> 100% methanol) followed by trituration with dichloromethane to give the title compound (56 mg, 17 % yield).
  • reaction mixture was concentrated and the residue was purified by flash column chromatography (25 g Snap cartridge, dichloromethane/ methanol-gradient, 20% -> 100% methanol) followed by trituration with dichloromethane to give the title compound (417 mg, 80 % yield).
  • the compounds of formula (I) of the present invention can be converted to any salt as described herein, by any method which is known to the person skilled in the art.
  • any salt of a compound of formula (I) of the present invention can be converted into the free compound, by any method which is known to the person skilled in the art.
  • compositions containing one or more compounds of the present invention can be utilised to achieve the desired pharmacological effect by administration to a patient in need thereof.
  • a patient for the purpose of this invention, is a mammal, including a human, in need of treatment for the particular condition or disease. Therefore, the present invention includes pharmaceutical compositions that are comprised of a pharmaceutically acceptable carrier and a pharmaceutically effective amount of a compound, or salt thereof, of the present invention.
  • a pharmaceutically acceptable carrier is preferably a carrier that is relatively non-toxic and innocuous to a patient at concentrations consistent with effective activity of the active ingredient so that any side effects ascribable to the carrier do not vitiate the beneficial effects of the active ingredient.
  • a pharmaceutically effective amount of compound is preferably that amount which produces a result or exerts an influence on the particular condition being treated.
  • the compounds of the present invention can be administered with pharmaceutically-acceptable carriers well known in the art using any effective conventional dosage unit forms, including immediate, slow and timed release preparations, orally, parenterally, topically, nasally, ophthalmically, optically, sublingually, rectally, vaginally, and the like.
  • the compounds can be formulated into solid or liquid preparations such as capsules, pills, tablets, troches, lozenges, melts, powders, solutions, suspensions, or emulsions, and may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions.
  • the solid unit dosage forms can be a capsule that can be of the ordinary hard- or soft-shelled gelatine type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and corn starch.
  • the compounds of this invention may be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatine, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, gum tragacanth, acacia, lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid, or magnesium, calcium or zinc stearate, dyes, colouring agents, and flavouring agents such as peppermint, oil of wintergreen, or cherry flavouring, intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient.
  • binders such as acacia, corn starch or gelatine
  • disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid
  • Suitable excipients for use in oral liquid dosage forms include dicalcium phosphate and diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent.
  • Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both.
  • Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example those sweetening, flavouring and colouring agents described above, may also be present.
  • the pharmaceutical compositions of this invention may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil such as liquid paraffin or a mixture of vegetable oils.
  • Suitable emulsifying agents may be (1 ) naturally occurring gums such as gum acacia and gum tragacanth, (2) naturally occurring phosphatides such as soy bean and lecithin, (3) esters or partial esters derived form fatty acids and hexitol anhydrides, for example, sorbitan monooleate, (4) condensation products of said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavouring agents.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil such as, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent such as, for example, beeswax, hard paraffin, or cetyl alcohol.
  • T e suspensions may also contain one or more preservatives, for example, ethyl or n-propyl p- hydro xybenzoate ; one or more colouring agents ; one or more flavouring agents ; and one or more sweetening agents such as sucrose or saccharin.
  • Syrups and elixirs may be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, and preservative, such as methyl and propyl parabens and flavouring and colouring agents.
  • the compounds of this invention may also be administered parenterally, that is, subcutaneously, intravenously, intraocularly, intrasynovially, intramuscularly, or interperitoneally, as injectable dosages of the compound in preferably a physiologically acceptable diluent with a pharmaceutical carrier which can be a sterile liquid or mixture of liquids such as water, saline, aqueous dextrose and related sugar solutions, an alcohol such as ethanol, isopropanol, or hexadecyl alcohol, glycols such as propylene glycol or polyethylene glycol, glycerol ketals such as 2,2-dimethyl-1 ,1 -dioxolane-4-methanol, ethers such as polyethylene glycol) 400, an oil, a fatty acid, a fatty acid ester or, a fatty acid glyceride, or an acetylated fatty acid glyceride, with or without the addition of a pharmaceutically acceptable surfactant
  • Suitable fatty acids include oleic acid, stearic acid, isostearic acid and myristic acid.
  • Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate.
  • Suitable soaps include fatty acid alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamine acetates ; anionic detergents, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates ; non-ionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and poly(oxyethylene-oxypropylene)s or ethylene oxide or propylene oxide copolymers ; and amphoteric detergents, for example, alkyl-beta-aminopropionates, and 2-alkylimidazoline quaternary ammonium salts, as well as mixtures.
  • suitable detergents include cationic detergents
  • compositions of this invention will typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimise or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile- lipophile balance (HLB) preferably of from about 12 to about 17. The quantity of surfactant in such formulation preferably ranges from about 5% to about 15% by weight.
  • the surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB.
  • surfactants used in parenteral formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • compositions may be in the form of sterile injectable aqueous suspensions.
  • suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents such as, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia ; dispersing or wetting agents which may be a naturally occurring phosphatide such as lecithin, a condensation product of an alkylene oxide with a fatty acid, for example, polyoxyethylene stearate, a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadeca- ethyleneoxycetanol, a condensation product of ethylene oxide with a partial ester derived form a fatty acid and a hexitol such as polyoxyethylene sorbitol monooleate, or a condensation product of an ethylene oxide with a partial ester derived from
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent.
  • Diluents and solvents that may be employed are, for example, water, Ringer's solution, isotonic sodium chloride solutions and isotonic glucose solutions.
  • sterile fixed oils are conventionally employed as solvents or suspending media.
  • any bland, fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid can be used in the preparation of injectables.
  • composition of the invention may also be administered in the form of suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are, for example, cocoa butter and polyethylene glycol.
  • transdermal delivery devices Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts.
  • the construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art (see, e.g. , US Patent No. 5,023,252, issued June 1 1 . 1991 , incorporated herein by reference).
  • patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • Controlled release formulations for parenteral administration include liposomal, polymeric microsphere and polymeric gel formulations that are known in the art.
  • a mechanical delivery device It may be desirable or necessary to introduce the pharmaceutical composition to the patient via a mechanical delivery device.
  • the construction and use of mechanical delivery devices for the delivery of pharmaceutical agents is well known in the art.
  • Direct techniques for, for example, administering a drug directly to the brain usually involve placement of a drug delivery catheter into the patient's ventricular system to bypass the blood-brain barrier.
  • One such implantable delivery system, used for the transport of agents to specific anatomical regions of the body is described in US Patent No. 5,01 1 ,472, issued April 30, 1991 .
  • compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired.
  • Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized. Such ingredients and procedures include those described in the following references, each of which is incorporated herein by reference: Powell, M. F. et al. , "Compendium of Excipients for Parenteral Formulations” PDA Journal of Pharmaceutical Science 6t Technology 1998, 52(5), 238-31 1 ; Strickley, R.G “Parenteral Formulations of Small Molecule Therapeutics Marketed in the United States (1999)-Part-1 " PDA Journal of Pharmaceutical Science & Technology 1999, 53(6), 324-349 ; and Nema, S. et al. , "Excipients and Their Use in Injectable Products” PDA Journal of Pharmaceutical Science & Technology 1997, 51 (4), 166-171 .
  • compositions for its intended route of administration include: acidifying agents (examples include but are not limited to acetic acid, citric acid, fumaric acid, hydrochloric acid, nitric acid) ;
  • alkali nizing agents examples include but are not limited to ammonia solution, ammonium carbonate, diethanolamine, monoethanolamine, potassium hydroxide, sodium borate, sodium carbonate, sodium hydroxide, triethanolamine, trolamine) ;
  • adsorbents examples include but are not limited to powdered cellulose and activated charcoal
  • aerosol propellents examples include but are not limited to carbon dioxide, CCI 2 F 2 , F 2 C C- CClFz and CCIF3
  • air displacement agents examples include but are not limited to nitrogen and argon
  • antifungal preservatives examples include but are not limited to benzoic acid, butylparaben, ethylparaben, methylparaben, propylparaben, sodium benzoate
  • benzoic acid examples include but are not limited to benzoic acid, butylparaben, ethylparaben, methylparaben, propylparaben, sodium benzoate
  • antimicrobial preservatives examples include but are not limited to benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetylpyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, phenylmercuric nitrate and thimerosal
  • examples include but are not limited to benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetylpyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, phenylmercuric nitrate and thimerosal
  • antioxidants examples include but are not limited to ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorus acid, monothioglycerol, propyl gallate, sodium ascorbate, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite) ;
  • binding materials examples include but are not limited to block polymers, natural and synthetic rubber, polyacrylates, polyurethanes, silicones, polysiloxanes and styrene- butadiene copolymers
  • buffering agents examples include but are not limited to potassium metaphosphate, dipotassium phosphate, sodium acetate, sodium citrate anhydrous and sodium citrate dihydrate
  • carrying agents examples include but are not limited to acacia syrup, aromatic syrup, aromatic elixir, cherry syrup, cocoa syrup, orange syrup, syrup, corn oil, mineral oil, peanut oil, sesame oil, bacteriostatic sodium chloride injection and bacteriostatic water for injection
  • examples include but are not limited to acacia syrup, aromatic syrup, aromatic elixir, cherry syrup, cocoa syrup, orange syrup, syrup, corn oil, mineral oil, peanut oil, sesame oil, bacteriostatic sodium chloride injection and bacteriostatic water for injection
  • chelating agents examples include but are not limited to edetate disodium and edetic acid
  • colourants examples include but are not limited to FD&C Red No. 3, FD&C Red No. 20, FD&C Yellow No. 6, FD&C Blue No. 2, D&C Green No. 5, D&C Orange No. 5, D&C Red No. 8, caramel and ferric oxide red
  • FD&C Red No. 3 examples include but are not limited to FD&C Red No. 3, FD&C Red No. 20, FD&C Yellow No. 6, FD&C Blue No. 2, D&C Green No. 5, D&C Orange No. 5, D&C Red No. 8, caramel and ferric oxide red
  • clarifying agents examples include but are not limited to bentonite
  • emulsifying agents examples include but are not limited to acacia, cetomacrogol, cetyl alcohol, glyceryl monostearate, lecithin, sorbitan monooleate, polyoxyethylene 50 monostearate) ;
  • encapsulating agents examples include but are not limited to gelatin and cellulose acetate phthalate
  • flavourants examples include but are not limited to anise oil, cinnamon oil, cocoa, menthol, orange oil, peppermint oil and vanillin
  • examples include but are not limited to anise oil, cinnamon oil, cocoa, menthol, orange oil, peppermint oil and vanillin
  • humectants examples include but are not limited to glycerol, propylene glycol and sorbitol
  • levigating agents examples include but are not limited to mineral oil and glycerin
  • oils examples include but are not limited to arachis oil, mineral oil, olive oil, peanut oil, sesame oil and vegetable oil) ;
  • ointment bases examples include but are not limited to lanolin, hydrophilic ointment, polyethylene glycol ointment, petrolatum, hydrophilic petrolatum, white ointment, yellow ointment, and rose water ointment
  • examples include but are not limited to lanolin, hydrophilic ointment, polyethylene glycol ointment, petrolatum, hydrophilic petrolatum, white ointment, yellow ointment, and rose water ointment
  • penetration enhancers include but are not limited to monohydroxy or polyhydroxy alcohols, mono-or polyvalent alcohols, saturated or unsaturated fatty alcohols, saturated or unsaturated fatty esters, saturated or unsaturated dicarboxylic acids, essential oils, phosphatidyl derivatives, cephalin, terpenes, amides, ethers, ketones and ureas
  • monohydroxy or polyhydroxy alcohols mono-or polyvalent alcohols
  • saturated or unsaturated fatty alcohols saturated or unsaturated fatty esters
  • saturated or unsaturated dicarboxylic acids saturated or unsaturated dicarboxylic acids
  • essential oils phosphatidyl derivatives
  • cephalin cephalin
  • terpenes amides, ethers, ketones and ureas
  • plasticizers examples include but are not limited to diethyl phthalate and glycerol
  • solvents examples include but are not limited to ethanol, corn oil, cottonseed oil, glycerol, isopropanol, mineral oil, oleic acid, peanut oil, purified water, water for injection, sterile water for injection and sterile water for irrigation
  • water for injection examples include but are not limited to diethyl phthalate and glycerol
  • solvents examples include but are not limited to ethanol, corn oil, cottonseed oil, glycerol, isopropanol, mineral oil, oleic acid, peanut oil, purified water, water for injection, sterile water for injection and sterile water for irrigation
  • stiffening agents examples include but are not limited to cetyl alcohol, cetyl esters wax, microcrystalline wax, paraffin, stearyl alcohol, white wax and yellow wax
  • stiffening agents include but are not limited to cetyl alcohol, cetyl esters wax, microcrystalline wax, paraffin, stearyl alcohol, white wax and yellow wax
  • suppository bases examples include but are not limited to cocoa butter and polyethylene glycols (mixtures)
  • surfactants examples include but are not limited to benzalkonium chloride, nonoxynol 10, oxtoxynol 9, polysorbate 80, sodium lauryl sulfate and sorbitan mono-palmitate
  • examples include but are not limited to cocoa butter and polyethylene glycols (mixtures)
  • surfactants examples include but are not limited to benzalkonium chloride, nonoxynol 10, oxtoxynol 9, polysorbate 80, sodium lauryl sulfate and sorbitan mono-palmitate
  • suspending agents examples include but are not limited to agar, bentonite, carbomers, carboxymethylcellulose sodium, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, kaolin, methylcellulose, tragacanth and veegum
  • agar bentonite
  • carbomers carboxymethylcellulose sodium, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, kaolin, methylcellulose, tragacanth and veegum
  • sweetening agents examples include but are not limited to aspartame, dextrose, glycerol, mannitol, propylene glycol, saccharin sodium, sorbitol and sucrose
  • sweetening agents include but are not limited to aspartame, dextrose, glycerol, mannitol, propylene glycol, saccharin sodium, sorbitol and sucrose
  • tablet anti-adherents examples include but are not limited to magnesium s tea rate and talc
  • tablet binders examples include but are not limited to acacia, alginic acid, carboxymethylcellulose sodium, compressible sugar, ethylcellulose, gelatin, liquid glucose, methylcellulose, non-crosslinked polyvinyl pyrrolidone, and pregelatinized starch
  • examples include but are not limited to acacia, alginic acid, carboxymethylcellulose sodium, compressible sugar, ethylcellulose, gelatin, liquid glucose, methylcellulose, non-crosslinked polyvinyl pyrrolidone, and pregelatinized starch
  • tablet and capsule diluents examples include but are not limited to dibasic calcium phosphate, kaolin, lactose, mannitol, microcrystalline cellulose, powdered cellulose, precipitated calcium carbonate, sodium carbonate, sodium phosphate, sorbitol and starch) ;
  • tablet coating agents examples include but are not limited to liquid glucose, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose, ethylcellulose, cellulose acetate phthalate and shellac) ;
  • tablet direct compression excipients examples include but are not limited to dibasic calcium phosphate
  • tablet disintegrants examples include but are not limited to alginic acid, carboxymethylcellulose calcium, microcrystalline cellulose, polacrillin potassium, cross- linked polyvinylpyrrolidone, sodium alginate, sodium starch glycollate and starch
  • examples include but are not limited to alginic acid, carboxymethylcellulose calcium, microcrystalline cellulose, polacrillin potassium, cross- linked polyvinylpyrrolidone, sodium alginate, sodium starch glycollate and starch
  • tablet glidants examples include but are not limited to colloidal silica, corn starch and talc) ;
  • tablet lubricants examples include but are not limited to calcium stearate, magnesium stearate, mineral oil, stearic acid and zinc stearate
  • examples include but are not limited to calcium stearate, magnesium stearate, mineral oil, stearic acid and zinc stearate
  • tablet/capsule opaquants examples include but are not limited to titanium dioxide
  • tablet polishing agents examples include but are not limited to carnuba wax and white wax
  • thickening agents examples include but are not limited to beeswax , cetyl alcohol and paraffin
  • tonicity agents examples include but are not limited to dextrose and sodium chloride
  • viscosity increasing agents examples include but are not limited to alginic acid, bentonite, carbomers, carboxymethylcellulose sodium, methylcellulose, polyvinyl pyrrolidone, sodium alginate and tragacanth
  • wetting agents examples include but are not limited to heptadecaethylene oxycetanol, lecithins, sorbitol monooleate, polyoxyethylene sorbitol monooleate, and polyoxyethylene stearate).
  • compositions according to the present invention can be illustrated as follows:
  • Sterile IV Solution A 5 mg/mL solution of the desired compound of this invention can be made using sterile, injectable water, and the pH is adjusted if necessary. The solution is diluted for administration to 1 - 2 mg/mL with sterile 5% dextrose and is administered as an IV infusion over about 60 min.
  • a sterile preparation can be prepared with (i) 100 - 1000 mg of the desired compound of this invention as a lyophilised powder, (ii) 32- 327 mg/mL sodium citrate, and (iii) 300 - 3000 mg Dextran 40.
  • the formulation is reconstituted with sterile, injectable saline or dextrose 5% to a concentration of 10 to 20 mg/mL, which is further diluted with saline or dextrose 5% to 0.2 - 0.4 mg/mL, and is administered either IV bolus or by IV infusion over 15 60 min.
  • Intramuscular suspension The following solution or suspension can be prepared, for intramuscular injection:
  • Hard Shell Capsules A large number of unit capsules are prepared by filling standard two- piece hard galantine capsules each with 100 mg of powdered active ingredient, 150 mg of lactose, 50 mg of cellulose and 6 mg of magnesium stearate.
  • Soft Gelatin Capsules A mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil is prepared and injected by means of a positive displacement pump into molten gelatin to form soft gelatin capsules containing 100 mg of the active ingredient. The capsules are washed and dried. The active ingredient can be dissolved in a mixture of polyethylene glycol, glycerin and sorbitol to prepare a water miscible medicine mix.
  • Tablets A large number of tablets are prepared by conventional procedures so that the dosage unit is 100 mg of active ingredient, 0.2 mg. of colloidal silicon dioxide, 5 mg of magnesium stearate, 275 mg of microcrystalline cellulose, 1 1 mg of starch, and 98.8 mg of lactose. Appropriate aqueous and non-aqueous coatings may be applied to increase palatability, improve elegance and stability or delay absorption.
  • Immediate Release Tablets/ Capsules These are solid oral dosage forms made by conventional and novel processes. These units are taken orally without water for immediate dissolution and delivery of the medication.
  • the active ingredient is mixed in a liquid containing ingredient such as sugar, gelatin , pectin and sweeteners. These liquids are solidified into solid tablets or caplets by freeze drying and solid state extraction techniques.
  • the drug compounds may be compressed with viscoelastic and thermoelastic sugars and polymers or effervescent components to produce porous matrices intended for immediate release, without the need of water.
  • a "fixed combination” in the present invention is used as known to persons skilled in the art and may be present as a fixed combination, a non-fixed combination or kit-of-parts.
  • a "fixed combination” in the present invention is used as known to persons skilled in the art and is defined as a combination wherein the said first active ingredient and the said second active ingredient are present together in one unit dosage or in a single entity.
  • a "fixed combination” is a pharmaceutical composition wherein the said first active ingredient and the said second active ingredient are present in admixture for simultaneous administration, such as in a formulation.
  • Another example of a "fixed combination” is a pharmaceutical combination wherein the said first active ingredient and the said second active ingredient are present in one unit without being in admixture.
  • a non-fixed combination or "kit-of-parts" in the present invention is used as known to persons skilled in the art and is defined as a combination wherein the said first active ingredient and the said second active ingredient are present in more than one unit.
  • a non-fixed combination or kit-of-parts is a combination wherein the said first active ingredient and the said second active ingredient are present separately.
  • the components of the non-fixed combination or kit-of-parts may be administered separately, sequentially, simultaneously, concurrently or chronologically staggered.
  • the compounds of this invention can be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutical agents where the combination causes no unacceptable adverse effects.
  • the present invention relates also to such combinations.
  • the compounds of this invention can be combined with known chemotherapeutic agents or anti-cancer agents, e.g. anti-hyper-proliferative or other indication agents, and the like, as well as with admixtures and combinations thereof.
  • Other indication agents include, but are not limited to, anti-angiogenic agents, mitotic inhibitors, alkylating agents, anti-metabolites, DNA-intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzyme inhibitors, toposisomerase inhibitors, biological response modifiers, or anti-hormones.
  • chemotherapeutic anti-cancer agents includes but is not limited to
  • the compounds of the invention may also be administered in combination with protein therapeutics.
  • protein therapeutics suitable for the treatment of cancer or other angiogenic disorders and for use with the compositions of the invention include, but are not limited to, an interferon (e.g. , interferon .alpha. , .beta. , or .gamma.
  • Monoclonal antibodies useful as the protein therapeutic include, but are not limited to, muromonab-CD3, abciximab, edrecolomab, daclizumab, gentuzumab, alemtuzumab, ibritumomab, cetuximab, bevicizumab, efalizumab, adalimumab, omalizumab, muromomab-CD3, rituximab, daclizumab, trastuzumab, palivizumab, basiliximab, and infliximab.
  • a compound of general formula (I ) as defined herein can optionally be administered in combination with one or more of the following: ARRY-1 62, ARRY-300, ARRY-704, AS- 703026, AZD-5363, AZD-8055, BEZ-235, BGT-226, B KM- 1 20, BYL-719, CAL- 101 , CC-223, CH- 51 32799, deforolimus, E-6201 , enzastaurin , GDC-0032, GDC-0068 , GDC-0623 , GDC-0941 , GDC-0973, GDC-0980, GSK-21 10183 , GSK-2126458, GSK-2141795, MK-2206, novolimus, OSI- 027, perifosine, PF-04691 502, PF-05212384, PX-866, rapamycin , RG-7167, RO-4987655, R0- 5126766, selumetinib, T
  • cytotoxic and/or cytostatic agents in combination with a compound or composition of the present invention will serve to:
  • a compound of the present invention may be used to sensitize a cell to radiation. That is, treatment of a cell with a compound of the present invention prior to radiation treatment of the cell renders the cell more susceptible to DNA damage and cell death than the cell would be in the absence of any treatment with a compound of the invention.
  • the cell is treated with at least one compound of the invention.
  • the present invention also provides a method of killing a cell, wherein a cell is administered one or more compounds of the invention in combination with conventional radiation therapy.
  • the present invention also provides a method of rendering a cell more susceptible to cell death, wherein the cell is treated with one or more compounds of the invention prior to the treatment of the cell to cause or induce cell death.
  • the cell is treated with at least one compound, or at least one method, or a combination thereof, in order to cause DNA damage for the purpose of inhibiting the function of the normal cell or killing the cell.
  • a cell is killed by treating the cell with at least one DNA damaging agent. That is, after treating a cell with one or more compounds of the invention to sensitize the cell to cell death, the cell is treated with at least one DNA damaging agent to kill the cell.
  • DNA damaging agents useful in the present invention include, but are not limited to, chemotherapeutic agents (e.g. , cisplatinum), ionizing radiation (X-rays, ultraviolet radiation), carcinogenic agents, and mutagenic agents.
  • a cell is killed by treating the cell with at least one method to cause or induce DNA damage.
  • methods include, but are not limited to, activation of a cell signalling pathway that results in DNA damage when the pathway is activated, inhibiting of a cell signalling pathway that results in DNA damage when the pathway is inhibited, and inducing a biochemical change in a cell, wherein the change results in DNA damage.
  • a DNA repair pathway in a cell can be inhibited, thereby preventing the repair of DNA damage and resulting in an abnormal accumulation of DNA damage in a cell.
  • a compound of the invention is administered to a cell prior to the radiation or other induction of DNA damage in the cell. In another aspect of the invention, a compound of the invention is administered to a cell concomitantly with the radiation or other induction of DNA damage in the cell. In yet another aspect of the invention, a compound of the invention is administered to a cell immediately after radiation or other induction of DNA damage in the cell has begun.
  • the cell is in vitro. In another embodiment, the cell is in vivo.
  • the compounds of the present invention have surprisingly been found to effectively inhibit tankyrases and may therefore be used for the treatment or prophylaxis of diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, or diseases which are accompanied with uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, particularly in which the uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses are affected by inhibition of tankyrases, such as, for example, haematological tumours, solid tumours, and/or metastases thereof, e.g.
  • leukaemias and myelodysplastic syndrome including leukaemias and myelodysplastic syndrome, malignant lymphomas, head and neck tumours including brain tumours and brain metastases, tumours of the thorax including non-small cell and small cell lung tumours, gastrointestinal tumours, endocrine tumours, mammary and other gynaecological tumours, urological tumours including renal, bladder and prostate tumours, skin tumours, and sarcomas, and/or metastases thereof.
  • the present invention covers a compound of general formula (I), or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, as described and defined herein, for use in the treatment or prophylaxis of a disease, as mentioned supra.
  • Another particular aspect of the present invention is therefore the use of a compound of general formula (I), described supra, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, for the prophylaxis or treatment of a disease.
  • Another particular aspect of the present invention is therefore the use of a compound of general formula (I) described supra or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, for manufacturing a pharmaceutical composition for the treatment or prophylaxis of a disease.
  • Another aspect of the present invention is the use of a compound of formula (I) or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, as described herein, in the manufacture of a medicament for the treatment or prophylaxis of a disease.
  • the diseases referred to in the four preceding paragraphs are diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, or diseases which are accompanied with uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, such as, for example, haematological tumours, solid tumours, and/or metastases thereof, e.g.
  • leukaemias and myelodysplastic syndrome including leukaemias and myelodysplastic syndrome, malignant lymphomas, head and neck tumours including brain tumours and brain metastases, tumours of the thorax including non-small cell and small cell lung tumours, gastrointestinal tumours, endocrine tumours, mammary and other gynaecological tumours, urological tumours including renal, bladder and prostate tumours, skin tumours, and sarcomas, and/or metastases thereof.
  • inappropriate within the context of the present invention, in particular in the context of "inappropriate cellular immune responses, or inappropriate cellular inflammatory responses", as used herein, is to be understood as meaning a response which is less than, or greater than normal, and which is associated with, responsible for, or results in, the pathology of said diseases.
  • the use is in the treatment or prophylaxis of diseases, wherein the diseases are haemotological tumours, solid tumours and/or metastases thereof.
  • Diseases further included in the context of the present invention are metabolic diseases (e.g. diabetes and obesity), fibrosis (e.g. lung fibrogenesis) and viral infection.
  • metabolic diseases e.g. diabetes and obesity
  • fibrosis e.g. lung fibrogenesis
  • the present invention relates to a method for using the compounds of the present invention and compositions thereof, to treat mammalian hyper-proliferative disorders.
  • Compounds can be utilized to inhibit, block, reduce, decrease, etc. , cell proliferation and/or cell division, and/or produce apoptosis.
  • This method comprises administering to a mammal in need thereof, including a human, an amount of a compound of this invention, or a pharmaceutically acceptable salt, isomer, polymorph, metabolite, hydrate, solvate or ester thereof ; etc. which is effective to treat the disorder.
  • Hyperproliferative disorders include but are not limited, e.g.
  • BPH benign prostate hyperplasia
  • solid tumours such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid and their distant metastases.
  • Those disorders also include lymphomas, sarcomas, and leukaemias.
  • breast cancer examples include, but are not limited to invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ.
  • cancers of the respiratory tract include, but are not limited to small-cell and non-small-cell lung carcinoma, as well as bronchial adenoma and pleuropulmonary blastoma.
  • brain cancers include, but are not limited to brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal tumour.
  • Tumours of the male reproductive organs include, but are not limited to prostate and testicular cancer.
  • Tumours of the female reproductive organs include, but are not limited to endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus.
  • Tumours of the digestive tract include, but are not limited to anal, colon, colorectal, oesophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers.
  • Tumours of the urinary tract include, but are not limited to bladder, penile, kidney, renal pelvis, ureter, urethral and human papillary renal cancers.
  • Eye cancers include, but are not limited to intraocular melanoma and retinoblastoma.
  • liver cancers include, but are not limited to hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma.
  • Skin cancers include, but are not limited to squamous cell carcinoma, Kaposi 's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer.
  • Head-and-neck cancers include, but are not limited to laryngeal, hypopharyngeal, nasopharyngeal, oropharyngeal cancer, lip and oral cavity cancer and squamous cell.
  • Lymphomas include, but are not limited to AlDS-related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Burkitt lymphoma, Hodgkin's disease, and lymphoma of the central nervous system.
  • Sarcomas include, but are not limited to sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma.
  • Leukemias include, but are not limited to acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia.
  • treating or “treatment” as stated throughout this document is used conventionally, e.g. , the management or care of a subject for the purpose of combating, alleviating, reducing, relieving, improving the condition of, etc. , of a disease or disorder, such as a carcinoma.
  • the present invention also provides methods of treating disorders and diseases associated with excessive and/or abnormal angiogenesis.
  • Inappropriate and ectopic expression of angiogenesis can be deleterious to an organism.
  • a number of pathological conditions are associated with the growth of extraneous blood vessels. These include, e.g. , diabetic retinopathy, ischemic retinal-vein occlusion, and retinopathy of prematurity [Aiello et al. New Engl. J. Med. 1994, 331 , 1 80 ; Peer et al. Lab. Invest. 1995, 72, 638], age-related macular degeneration [AMD ; see, Lopez et al. Invest. Opththalmol. Vis. Sci.
  • neovascular glaucoma neovascular glaucoma, psoriasis, retrolental fibroplasias, angiofibroma, inflammation, rheumatoid arthritis (RA), restenosis, in-stent restenosis, vascular graft restenosis, etc.
  • RA rheumatoid arthritis
  • the increased blood supply associated with cancerous and neoplastic tissue encourages growth, leading to rapid tumour enlargement and metastasis.
  • the growth of new blood and lymph vessels in a tumour provides an escape route for renegade cells, encouraging metastasis and the consequence spread of the cancer.
  • compounds of the present invention can be utilized to treat and/or prevent any of the aforementioned angiogenesis disorders, e.g. , by inhibiting and/or reducing blood vessel formation ; by inhibiting, blocking, reducing, decreasing, etc. endothelial cell proliferation or other types involved in angiogenesis, as well as causing cell death or apop
  • the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication.
  • the amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
  • the total amount of the active ingredient to be administered will generally range from about 0.001 mg/ kg to about 200 mg/kg body weight per day, and preferably from about 0.01 mg/kg to about 20 mg/ kg body weight per day.
  • Clinically useful dosing schedules will range from one to three times a day dosing to once every four weeks dosing.
  • "drug holidays" in which a patient is not dosed with a drug for a certain period of time may be beneficial to the overall balance between pharmacological effect and tolerability.
  • a unit dosage may contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered one or more times per day or less than once a day.
  • the average daily dosage for administration by injection will preferably be from 0.01 to 200 mg/ kg of total body weight.
  • the average daily rectal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight.
  • the average daily vaginal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight.
  • the average daily topical dosage regimen will preferably be from 0.1 to 200 mg administered between one to four times daily.
  • the transdermal concentration will preferably be that required to maintain a daily dose of from 0.01 to 200 mg/kg.
  • the average daily inhalation dosage regimen will preferably be from 0.01 to 100 mg/kg of total body weight.
  • the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like.
  • the desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
  • the diseases of said method are haematological tumours, solid tumour and/or metastases thereof.
  • the compounds of the present invention can be used in particular in therapy and prevention, i.e. prophylaxis, of tumour growth and metastases, especially in solid tumours of all indications and stages with or without pre-treatment of the tumour growth.
  • the average value also referred to as the arithmetic mean value
  • the median value represents the middle number of the group of values when ranked in ascending or descending order. If the number of values in the data set is odd, the median is the middle value. If the number of values in the data set is even, the median is the arithmetic mean of the two middle values.
  • Examples were synthesized one or more times. When synthesized more than once, data from biological assays represent average values or median values calculated utilizing data sets obtained from testing of one or more synthetic batch.
  • the potency of the compounds according to the invention was assessed by applying an in vitro inhibition assay.
  • the TNKS1 catalyzed NAD'-dependent ribosylation of a suitable protein substrate was detected using a commercially available biotin/streptavidin binding based assay format [TNKS1 Histone Ribosylation Assay Kit (Biotin-labeled NAD+), Catalog #80579; BPS Bioscience, San Diego, USA] .
  • biotin/streptavidin binding based assay format [TNKS1 Histone Ribosylation Assay Kit (Biotin-labeled NAD+), Catalog #80579; BPS Bioscience, San Diego, USA] .
  • the incorporation of a biotin-labeled NAD+ during the TNKS1 catalyzed ribosylation reaction was detected with a streptavidin-HRP coupled chemi-luminescent readout.
  • the intensity of the readout signal is proportional to the incorporated NAD'. Inhibition of TNKS1 leads to a decreased incorporation of NAD * and consequently to a lower readout signal intensity.
  • concentration of a test compound which inhibits the enzyme activity by 50% is reported as ICw.
  • the assay was conducted in a 384 well MTP format according to the manufacturer's protocol [http: / /www.bpsbioscience.com/poly-adp-ribose-polymerase/assay-kit/tnks1 - histone-ribosylation-assay-kit-biotin-labeled-nad-80579 referencing: Brown, J. A. , Marala, R. B. J. Pharmacol. Toxicol. Methods 2002 47: 137] and using a BMG Pherastar MTP reader [BMG-Labtech, Offenburg, Germany] .
  • TNKS1 Assay B The potency of selected compounds according to the invention was assessed applying a modified in vitro inhibition assay.
  • the TNKS1 catalyzed NAD+-dependent ribosylation of the enzyme itself (auto-parsylation) was detected using [ H]-NAD * as substrate and applying the scintillation proximity assay (SPA) method to detect tritium-labeled, parsylated TNKS1 .
  • the intensity of the readout signal is proportional to the incorporated [ 3 H]-NAD * .
  • Inhibition of TNKS1 leads to a decreased incorporation of [ 3 H]-NAD + and consequently to a lower readout signal intensity.
  • the concentration of a test compound which inhibits the enzyme activity by 50% is reported as IC».
  • TNKS1 enzyme sample was diluted with a modified assay buffer (50 mM MES pH 7.0, 1 mM DTT, 0.01 % Triton X-100) to a final concentration of 6 nM TNKS1 and 10x NAD * solution was diluted with the modified assay buffer (s. above) to a final 0.445x NAD' solution doped with 100 Bq/ ⁇ [ 3 H]-NAD + [Catalog #NET443H050UC, Perkin Elmer, Waltham , Massachusetts, USA ].
  • Substrate solution (10 ⁇ ) was incubated with different test compound concentrations (2.5 ul in 10 % DMSO in modified assay buffer) or control (2.5 ul 10 % DMSO in modified assay buffer only) and enzyme (10 ⁇ ) over night at room temperature.
  • the potency of the compounds according to the invention was assessed applying an in vitro inhibition assay.
  • the TNKS2 catalyzed NAD'-dependent ribosylation of a suitable protein substrate was detected using a commercially available biotin/streptavidin binding based assay format [TNKS2 Histone Ribosylation Assay Kit (Biotin-labeled NAD'), Catalog #80572; BPS Bioscience, San Diego, USA].
  • biotin/streptavidin binding based assay format [TNKS2 Histone Ribosylation Assay Kit (Biotin-labeled NAD'), Catalog #80572; BPS Bioscience, San Diego, USA].
  • the incorporation of a biotin-labeled NAD * during the TNKS2 catalyzed ribosylation reaction was detected with a streptavidin-HRP coupled chemi-luminescent readout.
  • the intensity of the readout signal is proportional to the incorporated NAD * .
  • Inhibition of TNKS2 leads to a decreased incorporation of NAD * and consequently to a lower readout signal intensity.
  • concentration of a test compound which inhibits the enzyme activity by 50% is reported as IC 3 ⁇ 4 o.
  • the assay was conducted in a 384 well MTP format according to the manufacturer's protocol [http: / /www. bpsbioscience.com/ poly-adp-ribose-polymerase/assay-kit/tnks2- histone-ribosylation-assay-kit-biotin-labeled-nad-80572 referencing: Brown, J. A. , Marala, R. B. J. Pharmacol. Toxicol. Methods 2002 47: 137] . and using a BMG Pherastar MTP reader [BMG-Labtech, Offenburg, Germany] .
  • the potency of selected compounds according to the invention was assessed applying a modified in vitro inhibition assay.
  • the TNKS2 catalyzed NAD'-dependent ribosylation of the enzyme itself was detected using [ 3 H] -NAD * as substrate and applying the scintillation proximity assay (SPA) method to detect tritium-labeled, parsylated TNKS2.
  • SPA scintillation proximity assay
  • the intensity of the readout signal is proportional to the incorporated [ 3 H] -NAD ⁇ I nhibition of TNKS2 leads to a decreased incorporation of [ 3 H]-NAD * and consequently to a lower readout signal intensity.
  • the concentration of a test compound which inhibits the enzyme activity by 50% is reported as IC».
  • TNKS2 enzyme sample was diluted with a modified assay buffer (50 mM MES pH 7.0, 1 mM DTT, 0.01 % Triton X-100) to a final concentration of 6 nM TNKS2 and 10x NAD' solution was diluted with the modified assay buffer (s. above) to a final 0.445x NAD * solution doped with 100 Bq/ ⁇ [ 3 H] -NAD + [Catalog #NET443H050UC, Perkin Elmer, Waltham , Massachusetts, USA] .
  • Substrate solution (10 ⁇ ) was incubated with different test compound concentrations (2.5 ⁇ in 10 % DMS0 in modified assay buffer) or control (2.5 ⁇ 10 % DMS0 in modified assay buffer only) and enzyme (10 ⁇ ) over night at room temperature.
  • Incorporated tritium was measured after addition of 50 ul SPA beads (1 mg/ ml) [Catalog #RPNQ0095 20 mg/ ml, Perkin Elmer, Waltham , Massachusetts, USA; diluted 1 : 10 with Dulbecco's phosphate buffered saline, PBS Catalog #08537, Sigma- Aldrich, Steinheim, Germany] and detection of the photon emission with a beta count plate reader [Wallac Micro Beta®, Perkin Elmer, Waltham , Massachusetts, USA].
  • the potency of the compounds according to the invention was assessed using a commercially available biotin/streptavidin binding assay kits from BPS Bioscience, San Diego, USA (Catalog #80551 ).
  • the incorporation of a biotin-labeled NAD' during the PARP1 catalyzed ribosylation of a suitable protein substrate was detected using with a streptavidin-HRP coupled chemi-luminescent readout.
  • the intensity of the readout signal is proportional to the incorporated NAD * .
  • Inhibition of PARP1 leads to a decreased incorporation of NAD' and consequently to a lower readout signal intensity.
  • the concentration of a test compound that inhibits the enzyme activity by 50% is reported as ICso-
  • the assay was conducted in a 96 well MTP format according to the manufacturer's protocol (Catalog No. 80551 ) and using a BMG Pherastar MTP reader [BMG-Labtech, Offenburg, Germany].
  • the potency of the compounds according to the invention was assessed using a commercially available biotin/streptavidin binding assay kits from BPS Bioscience, San Diego, USA (Catalog #80551 ).
  • the incorporation of a biotin-labeled NAD * during the PARP2 catalyzed ribosylation of a suitable protein substrate was detected using with a streptavidin-HRP coupled chemi-luminescent readout.
  • the intensity of the readout signal is proportional to the incorporated NAD'.
  • Inhibition of PARP2 leads to a decreased incorporation of NAD * and consequently to a lower readout signal intensity.
  • the concentration of a test compound that inhibits the enzyme activity by 50% is reported as ICso.
  • the assay was conducted in a 96 well MTP format according to the manufacturer's protocol (Catalog No. 80552) and using a BMG Pherastar MTP reader [BMG-Labtech, Offenburg, Germany]. 7 Cellular Assays
  • a cellular reporter assay was employed.
  • the corresponding assay cell was generated by transfection of the mammalian cell line HEK293 (ATCC, #CRL-1 73) with the Super TopFlash vector (Morin, Science 275, 1997, 1787-1790; Molenaar et al., Cell 86 (3), 1996, 391 -399).
  • the HEK293 cell line is cultivated at 37oC and 5% C02 in DMEM (Life Technologies, #41965-039), supplemented with 2 m glutamine, 20 mM HEPES, 1.4 mM pyruvate, 0.15% Na-bicarbonate and 10% foetal bovine serum (GIBCO, #10270). Stable transfectants were generated by selection with 300 g/ml Hygromycin.
  • HEK293 cells were cotransfected with the FOP control vector and pcDNA3.
  • the FOP vector is identical to the TOP construct, but it contains instead of functional TCF elements a randomized, non-functional sequence.
  • a stable transfected cell line was generated as well, based on selection with Geneticin (1 mg/ml).
  • the two cell lines were plated 24 h before beginning the test at 10000 cells per well in a 384 micro tit re plate (MTP) in 30 ⁇ growth medium.
  • MTP micro tit re plate
  • a dose response curve for the Wnt dependent luciferase expression was recorded by stimulating the assay cell line with human recombinant Wnt-3a (R&D, #5036- WN-010) at different concentrations for 16 h at 37 C and 5% CO2 followed by subsequent luciferase measurement, to determine the Wnt-3a EC50 for the HEK293 TOP cell line on the day of testing.
  • the recombinant human Wnt-3a was thereby applied between 2500 and 5 ng/ml in two-fold dilution steps.
  • the compounds were thereby serially prediluted in 100% DMSO and thereafter 50 fold into the CAFTY compound dilution buffer (described above). From this dilution 10 ⁇ were added in combination with the EC 50 concentration of recombinant Wnt3a to the cells in 30 ⁇ growth medium and incubated for 16 hours at 37 C and 5% CO ? .
  • luciferase assay buffer (1 : 1 mixture of luciferase substrate buffer (20 mM Tricine, 2.67 mM magnesium sulfate, 0.1 mM EDTA, 4 mM DTT, 270 ⁇ Coenzyme A, 470 ⁇ Luciferin, 530 ⁇ ATP, ph adjusted to pH 7.8 with a sufficient volume of 5M sodium hydroxide) and Triton buffer (30 ml Triton X-100, 1 15 ml glycerol, 308 mg Dithiothreitol, 4.45 g disodium hydrogen phosphate di hydrate, 3.03 g Tris .
  • HQ, ad 11 H?0, pH 7.8 was added in an equal volume to determine luciferase expression as a measure of Wnt signaling activity in a luminometer.
  • the Wnt inhibitory activity was determined as IC50 of resulting dose response curves.
  • SW403 cells (but not limited to) were seeded at 50000 cells per well in 96-well plates. After overnight incubation, cells were treated with testing compounds and vehicle at 37 C for 24 hours. Thereafter, cells were washed with PBS and then lysed in 15 ⁇ of lysis buffer (M-PER buffer, Thermo Scientific # 78505) with complete proteinase and phosphatase inhibitors (Roche, #11836153001 and # 04906837001 ). The lysates were centrifuged and the supernatants were harvested for analysis.
  • Tumor xenografts from in vivo studies were homogenized in a 2 ml tubes of Precellysl24 (Bertin Technologies, Villeurbanne, France) following with centrafugation to obtain tumor lysates.
  • Capillary electrophoresis-based Simple Western assays were carried out with Peggy SueTM NanoPro 1000 (ProteinSimple, California, USA).
  • the protein amounts of Axin2 (but not limited to) were detected using anti-Axin2 antibody (Cell Signaling, Catalog #2151 ), quantified using the area under the curve, and normalized against GAPDH (anti-GAPDH, Zytomed Systems GmbH, Catalog #RGM2-6C5, Berlin, Germany).
  • Real-time RT-PCR using a TaqMan fluorogenic detection system is a simple and sensitive assay for quantitative analysis of gene transcription.
  • the TaqMan fluorogenic detection system can monitor PCR in real time using a dual-labeled fluorogenic hybridization probe (TaqMan probe) and a polymerase with 5'-3' exonuclease activity.
  • TaqMan probe dual-labeled fluorogenic hybridization probe
  • a polymerase with 5'-3' exonuclease activity.
  • Cells from different cancer cell lines (as HCT116, but not limited to) were grown at 500- 1000 cells/well in 384 well cell culture plates. For cell lysis the cell medium was carefully removed. The cells were washed carefully once with 50 ⁇ /well PBS.
  • RT-PCR 2 ⁇ lysate (each) was transferred to a 384 well PCR plate.
  • the PCR reaction was composed by 5 ⁇ 2x One Step RT qPCR MasterMix Plus, 0.05 ⁇ Euroscript RT/RNAse Inhibitor (50 U/ ⁇ , 20 SJ/ ⁇ ) and 200 nM of the appropriate Primer/ Hydrolysis Probe mix (primer sequences of forward, reverse and probe are given below for each analysed gene of interest or house keeping gene). 10 ⁇ water were added per well. The plate was sealed with an adhesive optical film.
  • the RT-PCR protocol was setup with 30 min 48 C, then 10 min 95 C followed by 50 cycles of 15 sec 95 C/1 min 60 C and a cooling step of 40 C for 30 sec using a Lightcycler LS440 from Roche. Relative expression was calculated using CP values from the gene of interest (e.g. AXIN2, but not limited to) and a house keeping gene (L32).
  • the gene of interest e.g. AXIN2, but not limited to
  • L32 house keeping gene
  • AXIN2 forward primer: AGG CCAGTG AGTTG GTTGTC (SEQ ID NO. 4); reverse primer: AGCTCTGAGCCTTCAGCATC (SEQ ID NO. 5); probe: TCTGTGGGGAAGAAATTCCATACCG (SEQ ID NO. 6)
  • MTD maximal tolerable dose
  • the MTD is defined as the maximal applicable dose with a) no animal losing more than 10% body weight compared to initial body weight and b) no lethality during treatment phase.
  • test compounds were analysed in xenograft models on mice. Test compounds were dosed orally at their respective MTD as well as at sub-MTD dosages. In case the MTD could not be determined in previous MTD studies, the compounds were dosed at a maximum daily dose of 200 mg/kg (applied either in one single dose or split in 2 doses at 100 mg/ kg).
  • tumor cells 1 -5x10* tumor cells (suspended in 0.1 ml of 50% cell culture medium / 50% Matrigel) were subcutanously injected into the flank of each animal. Animals were randomized into treatment groups when tumors had reached an average area of 20-30 mm 2 and treatment was started. Body weight and tumor area of each animal were measured 2-3 times weekly, depending on tumor growth. Studies were terminated , when animals in the control groups (receiving only compound vehicle solutions) or treatment groups reached tumor areas - 1 50 mm 2 . At that time point, all groups in the study were terminated, tumors were isolated and weighed.
  • T/C Treatment/Control
  • RNA and protein were isolated from tumor samples following standard protocols.
  • Wnt/B-catenin target gene expression and Axin2 protein abundance were measured by standard qRT-PCR and Western blotting methods (see 7.2 and 7.3).
  • Table 1 IC3 ⁇ 4o values for selected examples in cellular HEK293 TOP and FOP assay as well as in TNKS1 and TNKS2 biochemical assay

Abstract

The present invention relates to compounds of general formula (I), in which X1, X2, R1, R2, R4, R5, R7, and R8 are as defined herein, to methods of preparing said compounds, to intermediate compounds useful for preparing said compounds, to pharmaceutical compositions and combinations comprising said compounds and to the use of said compounds for manufacturing a pharmaceutical composition for the treatment or prophylaxis of a disease, in particular of neoplasms, as a sole agent or in combination with other active ingredients.

Description

AMIDO-SUBSTITUTED AZOLE COMPOUNDS
The present invention relates to amido-substituted azole compounds of general formula (I) as described and defined herein, to methods of preparing said compounds, to intermediate compounds useful for preparing said compounds, to pharmaceutical compositions and combinations comprising said compounds and to the use of said compounds for manufacturing a pharmaceutical composition for the treatment or prophylaxis of a disease, in particular of neoplasms, as a sole agent or in combination with other active ingredients.
BACKGROUND OF THE INVENTION
Cancer is the leading cause of death in developed countries and the second leading cause of death in developing countries. Deaths from cancer worldwide are projected to continue rising, with an estimated 12 million deaths in 2030. While substantial progress has been made in developing effective therapies, there is a need for additional therapeutic modalities that target cancer and related diseases.
The complexity of cancer disease arises after a selection process for cells with acquired functional capabilities to enhance survival and/or resistance towards apoptosis and a limitless proliferative potential. In addition, bi-direction interaction of cancer cells and stromal cells provides further advantage of cancer cell survival and distant metastasis to the secondary organs and tissues [Liotta LA, Kohn EC. The m icroenvironmen t of the tumour- host interface. Nature 2001, 411:375]. Furthermore, cancer stem cells (CSCs) represent the apex in the hierarchical model of tumor genesis, heterogeneity and metastasis. CSCs possess the capacity for unlimited self-renewal, the ability to give rise to progeny cells, and also an innate resistance to cytotoxic therapeutics [Meacham CE and Morrison SJ. Tumour heterogeneity and cancer cell plasticity. Nature 2013 , 501 :328]. Thus, there is need to develop drugs for cancer therapy addressing distinct features of established tumors.
The discovery that Drosophila segment polarity gene Wingless had a common origin with the murine oncogene lnt-1 led to intensive studies on Wnt signaling pathway and identification of 19 mammalian Wnts and 10 Wnt receptors [Rijsewijk F, Schuermann M, Wagenaar E, Parren P, Weigel D, Nusse R. The Drosophila homolog of the mouse mammary oncogene int- 1 is identical to the segment polarity gene wingless. Cell. 1987, 50: 649]. Wnts are secreted glycoproteins which bind to cell surface receptors to initiate signaling cascades. Wnt signaling cascades have classified into two categories: canonical and non- canonical, differentiated by their dependence on β-catenin. Non-canonical Wnt pathways, such as the planar cell polarity (PCP) and Ca2+ pathway, function through β-catenin independent mechanisms. Canonical Wnt signalling is initiated when a Wnt ligand engages co- receptors of the Frizzled (Fzd) and low- density lipoprotein receptor related protein (LRP) families, ultimately leading to β-catenin stabilization, nuclear translocation and activation of target genes [Angers S, Moon RT. Proximal events in Wnt signal transduction. Nat Rev Mol Cell Biol. 2009, 10: 468. Cadigan KM, Liu Yl. Wnt signaling: complexity at the surface. J Cell Sci. 2006, 119: 395. Gordon MD, Nusse R. Wnt signaling: multiple pathways, multiple receptors, and multiple transcription factors. J Biol Chem. 2006, 281 : 22429. Huang H, He X. WntI beta- catenin signaling: new (and old) players and new insights. Curr Opin Cell Biol. 2008, 20: 119. Polakis P. The many ways of Wnt in cancer. Curr Opin Genet Dev. 2007, 17: 45. Rao TP, Kuhl M. An updated overview on Wnt signaling pathways: a prelude for more. Circ Res. 2010, 106: 1798].
In the absence of Wnt stimulus, β-catenin is held i n an inactive state by a multimeric "destruction" complex comprised of adenomatous polyposis coli (APC), Axin , glycogen synthase kinase 36 (GSK36) and casein kinase 1 a (CK1 a). APC and Axin function as a scaffold, permitting GSK36- and CK1 a-mediated phosphorylation of critical residues within β-catenin. These phosphorylation events mark β-catenin for ubiquitination recognition by the E3 ubiquitin ligase β-transducin-repeat-containing protein and lead to subsequent proteasomal degradation [He X, Semenov M, Tamai K, Zeng X. LDL receptor-related proteins 5 and 6 in Wnt/beta-catenin signaling: arrows point the way. Development.2004, 131 : 1663. Kimelman D, Xu W. beta- catenin destruction complex: insights and questions from a structural perspective. Oncogene 2006, 25: 7482 ].
In the presence of Wnt stimulus, Axin , GSK3B and Dvl are recruited to the co-receptor complex Fzd and LRP5/6 and lead to disruption of the β-catenin destruction complex. Therefore, β-catenin is stabilized and translocated to the nucleus. Once in the nucleus, 6- catenin forms a complex with members of the T-cell factor/lymphoid enhancer factor (TCF/ LEF) family of transcription factors, recruiting co-factors such as CBP, p300, TNIK, Bcl9 and Pygopus, and ultimately driving transcription of target genes including c-myc, 0ct4, cyclin D, survivin. [ Curtin JC and Lorenzi MV. Drug Discovery Approaches to Target Wnt Signaling in Cancer Stem Cells. Onco target 2010, 1 : 552]. Tankyrases play a key role in the destruction complex by regulating the stability of the rate-limiting AXIN proteins, RNF1 6 and tankyrase itself. The E3 ubiquitin ligase RNF1 6 recognizes tankyrase-mediated PARsylation and eartags AXIN, tankyrase and itself for proteasome-mediated degradation. Thus, tankyrases control the protein stability and turnover of key components of the destruction complex, and consequently the cellular levels of β-catenin [Huang SMA, Mishina YM, Liu S, Cheung A, Stegmeier F, et al. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature 2009, 461:614, Zhang Y, Liu S, Mickanin C, Feng Y, Charlat 0, et al. RNF146 is a poly(ADP-ribose)-directed E3 ligase that regulates axin degradation and Wnt signalling. Nature Cell Biology 2011, 13:623, 2011].
Aberrant regulation of the Wnt/B-catenin signaling pathway is a common feature across a broad spectrum of human cancers and evolves as a central mechanism in cancer biology. First of all, Wnt overexpression could lead to malignant transformation of mouse mammary tissue [Klaus A, BirchmeierW. Wnt signalling and its impact on development and cancer. Nat Rev Cancer 2008, 8: 387] . Second, tumor genome sequencing discovered the mutations in Wnt/B-catenin pathway components as well as epigenetic mechanisms that altered the expression of genes relevant to Wnt/ B-catenin pathway [Ying Y. et al. Epigenetic disruption of the WNT I beta-catenin signaling pathway in human cancers. Epigenetics 2009, 4:307] . Third, Wnt/B-catenin pathway also cooperates with other oncogenic signaling pathways in cancer and regulates tumorigenesis, growth, and metastasis [Klaus A, Birchmeier W. Wnt signalling and its impact on development and cancer. Nat Rev Cancer 8: 387-398, 2008]. In addition, there is an additional role of WNT signaling between tumor and stromal cell interaction leading to tumorigenesis and metastasis [Shahi P, Park D, Pond AC, Seethammagari M, Chiou S-H, Cho K, et al. Activation of Wnt signaling by chemically induced dimerization of LRP5 disrupts cellular homeostasis. PLoS ONE 2012, 7: e30814]. Furthermore, growing body of evidence indicates a critical role of β-catenin in CSCs [Eaves CJ, Humphries RK. Acute myeloid leukemia and the Wnt pathway. N Engl J Med. 2010, 362: 2326; Nusse R, Fuerer C, Ching W, Harnish K, Logan C, Zeng A, ten Berge D, Kalani Y. Wnt signaling and stem cell control. Cold Spring Harb Symp Quant Biol. 2008, 73 : 59; Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature 2005, 434: 843]. For example, stem- like colon cells with a high level of B-catenin signaling have a much greater tumorigenic potential than counterpart cells with low β-catenin signaling [Vermeulen L, De Sousa EMF, van der Heijden M, Cameron K, de Jong JH, Borovski T, Tuynman JB, Todaro M, Merz C, Rodermond H, Sprick MR, Kemper K, Richel DJ, Stassi G, Medema JP. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol. 2010, 12: 468]. Finally, activation of Wnt/6-catenin signalling pathway is also one of the major mechanism causing tumor recurrence and drug resistance. All these provide clear rationale to develop therapeutics targeting Wnt/6-catenin signaling pathway for the treatment of cancer.
One of the approaches to inhibit Wnt/6-catenin signaling pathway is to target druggable tankyrases. Tankyrase 1 (TNKS1 ) and tankyrase 2 (TNKS2) are poly(ADP-ribosyl)ases that are distinguishable from other members of the enzyme family by the structural features of the catalytic domain, and the presence of a sterile a-motif multimerization domain and an ankyrin repeat protein-interaction domain. Inhibition of TNKS blocks PARsylation of AXIN1 and AXIN2 and prevents their proteasomai degradation. As the consequence, TNKS inhibition enhances the activity of the β -catenin destruction complex and suppresses 6- catenin nuclear transclocation and the expression of β-catenin target genes.
In addition to its function in Wnt signaling through modulation of β-catenin destruction, tankyrases are also implicated in other cellular functions, including telomere homeostasis, mitotic spindle formation, vesicle transport linked to glucose metabolism, and viral replication. In these processes, tankyrases interact with target proteins, catalyze poly (ADP-ribosyl)ation, and regulate protein interactions and stability. For example, TNKS1 controls telomere homeostasis, which promotes telomeric extension by PARsylating TRF1. TRF1 is then targeted for proteasomai degradation by the E3 ubiquitin ligases F- box only protein 4 and/or RING finger LIM domain-binding protein (RLIM/RNF12), which facilitates telomere maintenance [Donigian JR and de Lange T. The role of the poly(ADP-ribose) polymerase tankyrase 1 in telomere length control by the TRF1 component of the shel terin complex. J Biol Chem 2007, 282:22662]. In addition, telomeric end-capping also requires canonical DNA repair proteins such as DNA-dependent protein kinase (DNAPK). TNKS1 stabilizes the catalytic subunit of DNAPK (DNAPKcs) by PARsylation [Dregalla RC, Zhou J, Idate RR, Battaglia CL, Liber HL, Bailey SM. Regulatory roles of tankyrase 1 at telomeres and in DNA repair: suppression of T-SCE and stabilization of DNA-PKcs. Aging 2010, 2(10):691] . Altered expression of TNKS1 and/or TNKS2, as well as genetic alterations in the tankyrase locus, have been detected in multiple tumors, e.g. fibrosarcoma, ovarian cancer, glioblastoma, pancreatic adenocarcinoma, breast cancer, astrocytoma, lung cancer, gastric cancer, and colon cancer [Lehti L, Chi N-W and Krauss S. Tankyrases as drug targets. FEBS Journal 2013, 280: 3576]. In addition, tankyrases appear to have impact on viral infections. For example, in HSV infection, it was shown that the virus cannot replicate efficiently in cells with depletion of both TNKS1 and TNKS2 [Li I, Yamauchi Y,
Kamakura M, Murayama T, Goshima F, Kimura H, Nishiyama Y, Herpes Simplex Virus Requires Poly(ADP-Ribose) Polymerase Activity for Efficient Replication and induces Extracellular Signal-Related Kinase- Dependen t Phosphorylation and ICPO-Dependent Nuclear Localization of Tankyrase 1. Journal of Virology 2012, 86(1): 492] .
Furthermore, a connection between tankyrases and glucose metabolism has been indicated. Thus, DNA polymorphism in a chromosomal region encoding tankyrase/ methionine sulfoxide reductase A is robustly associated with early-onset obesity. TNKS1 knockout mice appeared to have reduced fat pads, suggesting a potential connection of TNKS and obesity. TNKS may also play a role in tissue fibrosis.
In summary, tankyrases are promising drug targets in regulating WNT signaling, telomere length (e.g. telomere shortening and DNA damage induced cell death), lung fibrogenesis, myelination and viral infection. The invention presented here describes a novel class of tankyrase inhibitors and their potential clinical utility for the treatment of various diseases, such as cancer, aging, metabolic diseases (e.g. diabetes and obesity), fibrosis (e.g. lung fibrogenesis) and viral infection.
The following list of selected references relates to inhibitors of TNKS1 and/or TNKS2 described in the literature or in patents. However, the chemical structures and compound classes of the inhibitors described in these references are completely different from the chemical structures of the present invention:
Cancer Research 2013, 73 (10): 3132; J Med Chem 201 3, 56 (16): 6495; J Med Chem 2013, 56(3): 1341 ; J Med Chem 2013, 56(17): 7049; J Med Chem 201 3, 56(24): 10003; J Med Chem 2013, 56(7): 3012; J Med Chem 2013, 56(20): 7880; J Med Chem 2013, 56(1 1 ): 4320; ChemMedChem 2013, 8(12): 1978; ACS Med Chem Lett 2013, 4(12): 1 173; ACS Med Chem Lett 2013, 4(12): 1218; Acta Crystallogr Sect F Struct Biol Cryst Commun 2012, 68(Part 2): 1 15; J Med Chem 2012, 55(3): 1360; WO 2009059994, W0201 3164061 , W02014023390, WO 2012076898, WO 2013093508, WO 2013010092, WO 2013189905, WO 2013189865, WO 2013177349, WO 2013012723, WO 2013134079, WO 201 3182546, ACS Med Chem Lett, 2014, 6(3): 254, W02015150449. WO 2008/042283 (Exelixis) discloses imidazole-4,5-dicarboxamide derivatives as JAK2 modulators.
WO 2001 /000575 discloses heterocyclic dicarboxylic acid diamide derivatives as insecticides, including amido-substituted azole compounds.
However, the state of the art described above does not describe the specific substituted amido-substituted azole compounds of general formula (I) of the present invention as defined herein, i. e. an imidazole or an oxazole moiety, bearing :
- in its 4-position, a group of structure:
Figure imgf000007_0001
wherein :
* indicates the point of attachment of said groups with the rest of the molecule , and R' represents -OR9, or -N(R,0)Rn , which are as defined herein,
and
- in its 5-position, a group of structure:
Figure imgf000007_0002
wherein :
* indicates the point of attachment of said groups with the rest of the molecule, and
X2 represents CR6 or N, and R4, R5, R6, R7 and R8 are as defined herein, and
- in its 2-position, a substituent R2,
wherein :
R2 represents a group selected from hydrogen, d-Cralkyl, and CrCi-cycloalkyl ;
or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same, as described and defined herein, and as hereinafter referred to as "compounds of the present invention", or their pharmacological activity.
It has now been found, and this constitutes the basis of the present invention, that said compounds of the present invention have surprising and advantageous properties.
In particular, said compounds of the present invention have surprisingly been found to effectively inhibit TNKS1 and/or TNKS2 and may therefore be used for the treatment or prophylaxis of diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses or diseases which are accompanied with uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses mediated by TNKS1 and/or TNKS2 and/or mediated by the Wnt pathway, for example, haematological tumours, solid tumours, and/or metastases thereof, e.g. leukaemias and myelodysplastic syndrome, malignant lymphomas, head and neck tumours including brain tumours and brain metastases, tumours of the thorax including non-small cell and small cell lung tumours, gastrointestinal tumours, endocrine tumours, mammary and other gynaecological tumours, urological tumours including renal, bladder and prostate tumours, skin tumours, and sarcomas, and/or metastases thereof. Compounds of the present invention may additionally show improved selectivity for TNKS1 and/or TNKS2 (e.g. over other PARP (poly(ADP-ribose)-polymerase) enzymes), for the treatment of TNKS1 and/or TNKS2 driven diseases, by reaching sufficient efficacious dose without inducing toxicity driven by, for example, other PARPs inhibition. DESCRIPTION OF THE INVENTION
In accordance with a first aspect, the present invention covers compounds of general formula (I) :
Figure imgf000009_0001
in which :
X1 represents NR3 or 0,
X2 represents CR6 or N,
R1 represents a group selected from :
-OR9, and -N(R10)R11 ,
R2 represents a group selected from :
hydrogen, O-Cralkyl, and CrG-cycloalkyl,
R3 represents hydrogen,
R4 represents hydrogen,
R5 represents a group selected from :
hydrogen, and G-C3-alkyl,
R6 represents a group selected from :
hydrogen, and halogen,
R7 represents hydrogen,
R8 represents a group selected from :
aryl, aryl-(Ci-C4-alkyl)-, heteroaryl, and heteroaryl- (Ci-Gralkyl)-,
wherein aryl and heteroaryl groups are optionally substituted with one, two or three substituents, which are independently of each other selected from : G-G-alky , G-Gralkoxy, G-Grhydroxyalkyl, GrG-cycloalkyl, G-G-cycloalkoxy, G-Grhaloalkyl, G-G-haloalkoxy, halogen, cyano, nitro, hydroxy, -N(R'°)R" , R'°(R" )N-(G-G.-alkyl)-, R10(R11 )N-(C2-C4-alkoxy)-,
R9 represents G-Gralkyl,
R10 and R" are independently of each other selected from :
hydrogen, G -Cralkyl, G-G-cycloalkyl, (G-G-cycloalkyl)-(G-G-alkyl)-,
G-G-hydroxyalkyl, (G-G-alkoxy)-(G-G-alkyl)-, G-G-haloalkyl, H2N-(GrG-alkyl)- , (G -G- alkyl)N(H)(G-G-alkyl)-, (G-G-alkyl)2N(G-G-alkyl)-, 4-6 membered heterocycloalkyl, (4-6 membered heterocycloalkyl)- (G-G-alkyl)-,
wherein 4-6-membered heterocycloalkyl groups are optionally substituted with one or two substituents, which are independently of each other selected from :
G-alkyl, G-haloalkyl, G -alkoxy, G -hydroxyalkyl, G-haloalkoxy, halogen, and hydroxy; or,
R'° and R" together with the nitrogen atom to which they are attached form a 4-6-membered heterocycloalkyl group, in which one carbon atom is optionally replaced by a further heteroatom-containing group selected from NR'2, and 0, in which heterocycloalkyl group one additional ring atom is optionally replaced by C(=0), said 4-6-membered heterocycloalkyl group being optionally substituted with one or two substituents, which are independently of each other selected from :
G-alkyl, G-haloalkyl, G -alkoxy, G -haloalkoxy, halogen, and hydroxy;
R12 represents a group selected from :
hydrogen, and G-alkyl,
or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof , or a mixture of same. Definitions
Constituents which are optionally substituted as stated herein, may be substituted, unless otherwise noted, one or more times, independently from one another at any possible position. When any variable occurs more than one time in any constituent, each definition is independent.
When any variable occurs more than one time in any compound of general formula (I) as described herein, each definition is independent. For example, when R10, R11 , and/or R12 occur more than one time in any compound of formula (I ) each definition of R'°, R" , and R12 is independent.
A hyphen at the beginning or at the end of the constituent marks the point of attachment to the rest of the molecule. Should a ring be substituted the substitutent could be at any suitable position of the ring, also on a ring nitrogen atom if suitable.
The terms as mentioned in the present text have preferably the following meanings :
The term "comprising" when used in the specification includes "consisting of".
If it is referred to "as mentioned above" or "mentioned above" within the description it is referred to any of the disclosures made within the specification in any of the preceding pages.
If it is referred to "as mentioned herein", "described herein", "provided herein" or "stated herein" within the description it is referred to any of the disclosures made within the specification in any of the preceding or subsequent pages.
The term "halogen", "halogen atom", "halo-" or "Hal-" is to be understood as meaning a fluorine, chlorine, bromine or iodine atom.
The term "Ci-Ct-alkyl" is to be understood as preferably meaning a linear or branched, saturated, monovalent hydrocarbon group having 1 , 2, 3, or 4, carbon atoms, e.g. a methyl, ethyl, propyl, butyl, iso-propyl, iso-butyl, sec-butyl, tert-butyl group, more particularly 1 , 2 or 3 carbon atoms ("G -Cralkyl ") , e.g. a methyl, ethyl, n-propyl- or iso- propyl group, even more particularly 1 or 2 carbon atoms ("Ci -Cj-alkyl"), e.g. a methyl, ethyl group.
The term "C2-C4-alkyl" is to be understood as preferably meaning a linear or branched , saturated, monovalent hydrocarbon group having 2, 3 , or 4, carbon atoms, e.g. a ethyl, propyl, butyl, iso-propyl, iso-butyl, sec-butyl, tert-butyl group, more particularly 2 or 3 carbon atoms
Figure imgf000012_0001
e.g. a ethyl, n-propyl- or iso-propyl group, even more particularly 2 carbon atoms ("Cralkyl") , i. e. a ethyl group.
The term "G-Cs-hydroxya kyl" is to be understood as preferably meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "G -Cralkyl" is defined supra, and in which one or more hydrogen atoms is replaced by a hydroxy group, e.g. a hydroxymethyl, 1 -hydroxyethyl, 2-hydroxyethyl, 1 ,2-dihydroxyethyl, 3- hydroxypropyl , 2-hydroxypropyl, 2, 3-dihydroxypropyl, 1 , 3-dihydroxypropan-2-yl group.
The term "G-G-hydroxyalkyl" is to be understood as preferably meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "G-G-alkyl" is defined supra, and in which one or more hydrogen atoms is replaced by a hydroxy group, e.g. a 2-hydroxyethyl, 3-hydroxypropyl, 2-hydroxypropyl, 2, 3-dihydroxypropyl, 3-hydroxy- 2-methyl-propyl , 2-hydroxy-2-methyl-propyl group.
The term "CrC4-haloalkyl" is to be understood as preferably meaning a linear or branched , saturated, monovalent hydrocarbon group in which the term "G-G-alkyl" is defined supra, and in which one or more hydrogen atoms is replaced by a halogen atom , in identically or differently, i. e. one halogen atom being independent from another. Particularly, said halogen atom is F. Said G-G-ha oa kyl group is, for example, CF), -CHF2, -CH2F, -CF2CF3, - CH2CH2F, -CH2CHF2, -CH2CF3, or -CH2CH2CF3.
The term "G -G-alkoxy" is to be understood as preferably meaning a linear or branched, saturated , monovalent, hydrocarbon group of formula 0-alkyl having 1 , 2 , or 3 carbon atoms, in which the term "alkyl" is defined supra, e.g. a methoxy, ethoxy, n-propoxy, or iso-propoxy group, or an isomer thereof.
The term "G-Crhaloalkoxy" is to be understood as preferably meaning a linear or branched, saturated, monovalent G-Ci-alkoxy group, as defined supra, in which one or more of the hydrogen atoms is replaced, in identically or differently, by a halogen atom. Particularly, said halogen atom is F. Said G-Crhaloalkoxy group is, for example, OCF3, - OCHF?, -OCH7F, -OCF2CF3) or -OCH2CF3. The term "C3-C -cycloalkyl" is to be understood as meaning a saturated, monovalent, monocyclic hydrocarbon ring which contains 3, or 4, carbon atoms ("CrG-cycloalkyl"). Said G-G-cyc oa ky group is for example, a monocyclic hydrocarbon ring, e.g. a cyclopropyl, or cyclobutyl ring. The term "C3-G-cycloalkoxy" is to be understood as preferably meaning a saturated, monovalent, hydrocarbon ring which contains 3. or 4 carbon atoms of formula 0- cycloalkyl, in which the term "cycloalkyl" is defined supra, e.g. a cyclopropyloxy, or cyclobutyloxy.
The term "4- to 6-membered heterocycloalkyl", is to be understood as meaning a saturated, monovalent, monocyclic hydrocarbon ring which contains 3, 4 or 5 carbon atoms and a heteroatom-containing group selected from N, NR12, 0, S,
Figure imgf000013_0001
wherein one carbon atom is optionally replaced by a further heteroatom-containing group selected from NR12, 0, S, S(=0) and S(=0)?, in which R12 represents a hydrogen atom, or a G-alkyl- group, and in which heterocycloalkyl group one additional ring atom is optionally replaced by C(=0); it being possible for said heterocycloalkyl group to be attached to the rest of the molecule via any one of the carbon atoms or, if present, the nitrogen atom. Particularly, said 4- to 6-membered heterocycloalkyl contains 3, 4 or 5 carbon atoms and a heteroatom- containing group selected from N, NR'2, 0, S, S(=0) and S(=0 , wherein one carbon atom is optionally replaced by a further heteroatom-containing group selected from NR12, and 0, in which R12 represents a hydrogen atom, or a G-alkyl- group, and in which heterocycloalkyl group one additional ring atom is optionally replaced by C(=0). A heteroatom-containing group as defined herein is to be understood as meaning a group containing a heteroatom , such as NR'2, S(=0) and S(=0)?, or a single heteroatom such as N, 0 and S. Particularly, without being limited thereto, said heterocycloalkyl can be a 4-membered ring, such as an azetidinyl, oxetanyl, or a 5-membered ring, such as tetrahydrofuranyl, dioxolinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, or a 6-membered ring, such as tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, or N- methylpiperazinyl. Optionally, said heterocycloalkyl can be benzo fused. Particularly, without being limited thereto, 4- to 6-membered heterocycloalkyl can be selected from piperazinyl, tetrahydro-2H-pyranyl, tetrahydrofuranyl, pyrrolidinyl, piperidinyl, morpholinyl, azetidinyl, 2-oxoimidazolidinyl, 2-oxopyrrolidinyl and 1 , 1 - dioxidothiomorpholinyl. More particularly, without being limited thereto, 4- to 6- membered heterocycloalkyl can be selected from piperazin-1 -yl, tetrahydro-2H-pyran-4-yl, tetrahydrofuran-3-yl, pyrrolidin-1 -yl, pyrrolidin-2-yl, pyrrolidin-3-yl, piperidin-4-yl, piperidin-1 -yl, piperidin-2-yl, piperidin-3-yl, morpholin-4-yl, azetidin-1 -yl, tetrahydrofuran-2-yl, 2-oxoimidazolidin-1 -yl, 2-oxopyrrolidin-1 -yl and 1 , 1 - dioxidothiomorpholin-4-yl. The term "aryl" is to be understood as preferably meaning a monovalent, aromatic or partially aromatic, mono- or bicyclic hydrocarbon ring having 6, 7, 8, 9 or 10 carbon atoms (a "C6-Cio-aryl" group), particularly a ring having 6 carbon atoms (a "Ce-aryl" group), e.g. a phenyl group; or a ring having 9 carbon atoms (a "Cg-aryl" group), e.g. an indanyl or indenyl group, or a ring having 10 carbon atoms (a "Cio-aryl" group), e.g. a tetralinyl, dihydronaphthyl, or naphthyl group. In a preferred embodiment aryl is phenyl.
The term "heteroaryl" is understood as preferably meaning a monovalent, monocyclic aromatic ring system having 5 or 6 ring atoms (a "5- to 6-membered heteroaryl" group), which contains at least one heteroatom which may be identical or different, said heteroatom being such as oxygen, nitrogen, NH or sulfur. Particularly, heteroaryl is selected from thienyl, furanyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl etc. , or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, etc. More particularly, without being limited thereto, heteroaryl can be selected from pyrazolyl, thienyl, pyridyl, furanyl, thiazolyl, oxazolyl, and pyrazinyl.
In general, and unless otherwise mentioned, the heteroarylic or heteroarylenic radicals include all the possible isomeric forms thereof, e.g. the positional isomers thereof. Thus, for some illustrative non-restricting example, the term pyridinyl or pyridinylene includes pyridin-2-yl, pyridin-2-ylene, pyridin-3-yl, pyridin-3-ylene, pyridin-4-yl and pyridin-4-ylene; or the term thienyl or thienylene includes thien-2-yl, thien-2-ylene, thien-3-yl and thien-3- ylene.
In general, and unless otherwise mentioned, the heteroarylic radicals include all the possible isomeric forms thereof, e.g. the positional isomers thereof. Thus, for some illustrative non-restricting example, the term pyridinyl includes pyridin-2-yl, pyridin-3-yl and pyridin-4-yl.
In general, and unless otherwise mentioned, aromatic and non-aromatic (hetero)cyclic groups, may optionally be substituted as defined herein. The substituents may be present both when said aromatic and non-aromatic (hetero)cyclic groups exist as a (unitary) constituent, such as, for example, G-G-cycloalkyl, 4- to 6-membered heterocycloalkyl, aryl and heteroaryl groups, or as part of a constituent composed of more than one part, such as, for example, (G-G-cycloalkyl)-G-G-alkyl-, (4- to 6-membered heterocycloalkyl)- (CrC -alkyl)- , aryl-(G-G-alkyl)-, and heteroaryl- (C i -G-alkyl )- , for example. The present invention includes all suitably substituted aromatic and non-aromatic (hetero)cyclic groups both as a (unitary) constituent, or as part of a constituent composed of more than one part. In this context "suitably" is to be understood as meaning chemically possible to be made by methods within the knowledge of a skilled person.
The term "Ci-G.", as used throughout this text, e.g. in the context of the definition of "G- G-alkyl", or "G-G-haloalkyl", is to be understood as meaning an alkyl group having a finite number of carbon atoms of 1 to 4, i.e. 1 , 2, 3, or 4 carbon atoms. It is to be understood further that said term "G-G," is to be interpreted as any sub-range comprised therein, e.g. G-G. , G- , G-G , G-G , G-G ; particularly G-G , G-G , G-G. Similarly, as used herein, the term "C2-C4", as used throughout this text, e.g. in the context of the definitions of "C2-C4-alkyl", and "C2-C4-hydroxyalkyl" is to be understood as meaning an a Iky I group or a hydroxyalkyl group having a finite number of carbon atoms of 2 to 4, i. e. 2, 3, or 4 carbon atoms. It is to be understood further that said term "C2-C4" is to be interpreted as any sub-range comprised therein, e.g. C2-C4 , C3-C4 , C2-C3; particularly
The term "substituted" means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded , and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
The term "optionally substituted" means optional substitution with the specified groups, radicals or moieties.
Ring system substituent means a substituent attached to an aromatic or nonaromatic ring system which, for example, replaces an available hydrogen on the ring system.
As used herein, the term "one or more", e.g. in the definition of the substituents of the compounds of the general formulae of the present invention, is understood as meaning "one, two, three, four or five, particularly one, two, three or four, more particularly one, two or three, even more particularly one or two". The invention also includes all suitable isotopic variations of a compound of the invention. An isotopic variation of a compound of the invention is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually or predominantly found in nature. Examples of isotopes that can be incorporated into a compound of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine, chlorine, bromine and iodine, such as H (deuterium), 3H (tritium), "C, 13C, 14C, 15N, ,70, 180, 32P, 33P, 3S, 34S, 35S, 36S, 18F, 6Cl, 82Br, 123l, 124l, 125l, 129l and 131 l, respectively. Certain isotopic variations of a compound of the invention, for example, those in which one or more radioactive isotopes such as 3H or 14C are incorporated , are useful in drug and/or substrate tissue distribution studies. Tritiated and carbon-14, i.e. , 14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence is preferred in some circumstances. Isotopic variations of a compound of the invention can generally be prepared by conventional procedures known by a person skilled in the art such as by the illustrative methods or by the preparations described in the examples hereafter using appropriate isotopic variations of suitable reagents.
Where the plural form of the word compounds, salts, polymorphs, hydrates, solvates and the like, is used herein , this is taken to mean also a single compound , salt, polymorph , isomer, hydrate, solvate or the like.
By "stable compound' or "stable structure" is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
The compounds of this invention optionally contain one or more asymmetric centre, depending upon the location and nature of the various substituents desired. Asymmetric carbon atoms is present in the (R) or (S) configuration , resulting in racemic mixtures in the case of a single asymmetric centre, and diastereomeric mixtures in the case of multiple asymmetric centres. I n certain instances, asymmetry may also be present due to restricted rotation about a given bond , for example, the central bond adjoining two substituted aromatic rings of the specified compounds.
The compounds of the present invention optionally contain sulphur atoms which are asymmetric, such as an asymmetric sulfoxide, of structure:
Figure imgf000017_0001
, for example,
in which * indicates atoms to which the rest of the molecule can be bound. Substituents on a ring may also be present in either cis or trans form. It is intended that all such configurations (including enantiomers and diastereomers), are included within the scope of the present invention.
Preferred compounds are those which produce the more desirable biological activity. Separated, pure or partially purified isomers and stereoisomers or racemic or diastereomeric mixtures of the compounds of this invention are also included within the scope of the present invention. The purification and the separation of such materials can be accomplished by standard techniques known in the art.
The optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example, by the formation of diastereoisomeric salts using an optically active acid or base or formation of covalent diastereomers. Examples of appropriate acids are tartaric, diacetyl tartaric, ditoluoyltartaric and camphorsulfonic acid. Mixtures of diastereoisomers can be separated into their individual diastereomers on the basis of their physical and/or chemical differences by methods known in the art, for example, by chromatography or fractional crystallisation. The optically active bases or acids are then liberated from the separated diastereomeric salts. A different process for separation of optical isomers involves the use of chiral chromatography (e.g. , chiral HPLC columns), with or without conventional derivatisation, optimally chosen to maximise the separation of the enantiomers. Suitable chiral HPLC columns are manufactured by Daicel, e.g. , Chiracel OD and Chiracel OJ among many others, all routinely selectable. Enzymatic separations, with or without derivatisation, are also useful. The optically active compounds of this invention can likewise be obtained by chiral syntheses utilizing optically active starting materials.
In order to limit different types of isomers from each other reference is made to lUPAC Rules Section E (Pure Appl Chem 45, 1 1 -30, 1976).
The present invention includes all possible stereoisomers of the compounds of the present invention as single stereoisomers, or as any mixture of said stereoisomers, e.g. R- or S- isomers, or E- or Z-isomers, in any ratio. Isolation of a single stereoisomer, e.g. a single enantiomer or a single diastereomer, of a compound of the present invention is achieved by any suitable state of the art method , such as chromatography, especially chiral chromatography, for example. Further, the compounds of the present invention may exist as tautomers. For example, any compound of the present invention which contains a pyrazole moiety as a heteroaryl group for example can exist as a 1 H tautomer, or a 2H tautomer, or even a mixture in any amount of the two tautomers, namely :
Figure imgf000019_0001
1 H-tautomer 2H-tautomer
Particularly, when X1 represents NR\ wherein R3 represents a hydrogen atom , the present invention can exist as one of the below tautomers, or even in a mixture in any amount of the two tautomers, namely:
Figure imgf000019_0002
0 ; 0
The present invention includes all possible tautomers of the compounds of the present invention as single tautomers, or as any mixture of said tautomers, in any ratio.
Further, the compounds of the present invention can exist as N-oxides, which are defined in that at least one nitrogen of the compounds of the present invention is oxidised. The present invention includes all such possible N-oxides.
The present invention also relates to useful forms of the compounds as disclosed herein, such as metabolites, hydrates, solvates, prodrugs, salts, in particular pharmaceutically acceptable salts, and co-precipitates.
The compounds of the present invention can exist as a hydrate, or as a solvate, wherein the compounds of the present invention contain polar solvents, in particular water, methanol or ethanol for example as structural element of the crystal lattice of the compounds. The amount of polar solvents, in particular water, may exist in a stoichiometric or non-stoichiometric ratio. In the case of stoichiometric solvates, e.g. a hydrate, hemi-, (semi-), mono- , sesqui-, di- , tri- , tetra- , penta- etc. solvates or hydrates, respectively, are possible. The present invention includes all such hydrates or solvates.
Further, the compounds of the present invention can exist in free form, e.g. as a free base, or as a free acid, or as a zwitterion, or can exist in the form of a salt. Said salt may be any salt, either an organic or inorganic addition salt, particularly any pharmaceutically acceptable organic or inorganic addition salt, customarily used in pharmacy.
The term "pharmaceutically acceptable salt" refers to a relatively non-toxic, inorganic or organic acid addition salt of a compound of the present invention. For example, SCO S. M. Berge, er al. "Pharmaceutical Salts, " J. Pharm. Sci. 1977, 66, 1 -19.
A suitable pharmaceutically acceptable salt of the compounds of the present invention may be, for example, an acid-addition salt of a compound of the present invention bearing a nitrogen atom, in a chain or in a ring, for example, which is sufficiently basic, such as an acid-addition salt with an inorganic acid, such as hydrochloric, hydrobromic, hydroiodic, sulfuric, bi sulfuric, phosphoric, or nitric acid, for example, or with an organic acid, such as formic, acetic, acetoacetic, pyruvic, trifluoroacetic, propionic, butyric, hexanoic, heptanoic, undecanoic, lauric, benzoic, salicylic, 2-(4-hydroxybenzoyl)-benzoic, camphoric, cinnamic, cyclopentanepropionic, digluconic, 3-hydroxy-2-naphthoic, nicotinic, pamoic, pectinic, persulfuric, 3-phenylpropionic, picric, pivalic, 2-hydroxyethanesulfonate, itaconic, sulfamic, trifluoromethanesulfonic, dodecylsulfuric, ethansulfonic, benzenesulfonic, para-toluenesulfonic, methansulfonic, 2-naphthalenesulfonic, naphthalinedisulfonic, camphorsulfonic acid, citric, tartaric, stearic, lactic, oxalic, malonic, succinic, malic, adipic, alginic, maleic, fumaric, D-gluconic, mandelic, ascorbic, glucoheptanoic, glycerophosphoric, aspartic, sulfosalicylic, hemisulfuric, or thiocyanic acid, for example.
Further, another suitably pharmaceutically acceptable salt of a compound of the present invention which is sufficiently acidic, is an alkali metal salt, for example a sodium or potassium salt, an alkaline earth metal salt, for example a calcium or magnesium salt, an ammonium salt or a salt with an organic base which affords a physiologically acceptable cation, for example a salt with N-methyl-glucamine, dimethyl-glucamine, ethyl-glucamine, lysine, dicyclohexylamine, 1 ,6-hexadiamine, ethanolamine, glucosamine, sarcosine, serinol, tris-hydroxy-methyl-aminomethane, aminopropandiol, sovak-base, 1 -amino-2, 3,4- butantriol. Additionally, basic nitrogen containing groups may be quaternised with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides ; dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate ; and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and strearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.
Those skilled in the art will further recognise that acid addition salts of the claimed compounds may be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods. Alternatively, alkali and alkaline earth metal salts of acidic compounds of the invention are prepared by reacting the compounds of the invention with the appropriate base via a variety of known methods.
The present invention includes all possible salts of the compounds of the present invention as single salts, or as any mixture of said salts, in any ratio.
In the present text, in particular in the Experimental Section, for the synthesis of intermediates and of examples of the present invention, when a compound is mentioned as a salt form with the corresponding base or acid, the exact stoichiometric composition of said salt form, as obtained by the respective preparation and/or purification process, is, in most cases, unknown.
Unless specified otherwise, suffixes to chemical names or structural formulae such as "hydrochloride", "trifluoroacetate", "sodium salt", or "x HCl", "x CF3COOH", "x Na*", for example, are to be understood as not a stoichiometric specification, but solely as a salt form.
This applies analogously to cases in which synthesis intermediates or example compounds or salts thereof have been obtained, by the preparation and/or purification processes described, as solvates, such as hydrates with (if defined) unknown stoichiometric composition.
As used herein, the term "in vivo hydrolysable ester" is understood as meaning an in vivo hydrolysable ester of a compound of the present invention containing a carboxy or hydroxy group, for example, a pharmaceutically acceptable ester which is hydrolysed in the human or animal body to produce the parent acid or alcohol. Suitable pharmaceutically acceptable esters for carboxy include for example alkyl, cycloalkyl and optionally substituted phenylalkyl, in particular benzyl esters, G-Ce alkoxymethyl esters, e.g. methoxymethyl, G-Ce alkanoyloxymethyl esters, e.g. pivaloyloxymethyl, phthalidyl esters, C3-C8 cycloalkoxy-carbonyloxy-CrG> alkyl esters, e.g. 1 -cyclohexylcarbonyloxyethyl ; 1 , 3- dioxolen-2-onylmethyl esters, e.g. 5-methyl-1 ,3-dioxolen-2-onylmethyl ; and G -Gs- alkoxycarbonyloxyethyl esters, e.g. 1 -methoxycarbonyloxyethyl, and may be formed at any carboxy group in the compounds of this invention.
An in vivo hydrolysable ester of a compound of the present invention containing a hydroxy group includes inorganic esters such as phosphate esters and [alpha] -acyloxyalkyl ethers and related compounds which as a result of the in vivo hydrolysis of the ester breakdown to give the parent hydroxy group. Examples of [alpha] -acyloxyalkyl ethers include acetoxymethoxy and 2,2-dimethylpropionyloxymethoxy. A selection of in vivo hydrolysable ester forming groups for hydroxy include alkanoyl, benzoyl, phenylacetyl and substituted benzoyl and phenylacetyl, alkoxycarbonyl (to give alkyl carbonate esters), dialkylcarbamoyl and N-(dialkylaminoethyl)-N-alkylcarbamoyl (to give carbamates), dialkylaminoacetyl and carboxyacetyl. The present invention covers all such esters.
Furthermore, the present invention includes all possible crystalline forms, or polymorphs, of the compounds of the present invention, either as single polymorph, or as a mixture of more than one polymorph, in any ratio.
In accordance with a second aspect, the present invention covers compounds of general formula (I), supra, in which :
X1 represents NR;t,
X2 represents CR6,
R' represents a group selected from :
-OR9, and -N(R10)R11 ,
R2 represents a group selected from :
hydrogen, and G-alkyl,
R3 represents hydrogen,
R4 represents hydrogen, R5 represents a group selected from :
hydrogen, and G-a ky ,
R6 represents a group selected from :
hydrogen, and fluorine,
R7 represents hydrogen,
R8 represents a group selected from :
aryl, aryl-(CrC4-alkyl)-, heteroaryl, and heteroaryl- (Ci -C4-alkyl)-,
wherein aryl and heteroaryl groups are optionally substituted with one, two or three substituents, which are independently of each other selected from :
G-G-alkyl, G-Cs-a koxy, G-Cs-hydroxya kyl, G-Grhaloalkyl, halogen, cyano, -N(R10)R11 ,
R9 represents G-G-alkyl,
R'° and R" are independently of each other selected from :
hydrogen, G-G-alkyl, C3 -cycloalkyl, (C3-C4-cycloalkyl)-(Ci -C3-alkyl)-,
G-Grhydroxyalkyl, (Ci-alkoxy)- (C2-C3-alkyl)-, G-G-ha oalky , HjN-iGi-G-alkyl)-, (G- alkyl)N(H)(G-G-alkyl)-, (G-alkyl)2N(G-G-alkyl)-, (4-6 membered heterocycloalkyl)- (G-G- alkyl)-, wherein 4-6-membered heterocycloalkyl groups are optionally substituted with one or two substituents, which are independently of each other selected from :
G-alkyl, G-haloalkyl, G-alkoxy, G-hydroxyalky , G-haloalkoxy, halogen, and hydroxy; or,
R10 and Rn together with the nitrogen atom to which they are attached form a 4-6-membered heterocycloalkyl group, in which one carbon atom is optionally replaced by a further heteroatom-containing group selected from NR12, and 0, in which heterocycloalkyl group one additional ring atom is optionally replaced by C(=0), said 4-6-membered heterocycloalkyl group being optionally substituted with one or two substituents, which are independently of each other selected from :
G-alkyl, G-haloalkyl, G-alkoxy, G-haloalkoxy, halogen, and hydroxy;
R'2 represents a group selected from :
hydrogen, and G-alkyl, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.
In accordance with a third aspect, the present invention covers compounds of general formula (I ), supra, in which :
X1 represents NR3,
X2 represents CR6,
R1 represents -N (R, 0)R" ,
R2 represents hydrogen ,
R3 represents hydrogen ,
R4 represents hydrogen,
R5 represents hydrogen,
R0 represents hydrogen,
R7 represents hydrogen,
R8 represents a group selected from
aryl, aryl-(Ci -G-alkyl)-,
wherein aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
halogen ,
R'° and R11 are independently of each other selected from :
hydrogen , Ci -G-alkyl, C3-C4-cycloalkyl, (C3-C4-cycloalkyl)-(Ci -alkyl)-,
CrCrhydroxyalkyl, (Ci -alkoxy)- (C?-C3-alkyl)-, CrC3-haloalkyl, HjN- iCralkyl)- , (G - alkyl)N(H )(C2-alkyl)-, (Ci-alkyl)2N(C2-alkyl)-, (6-membered heterocycloalkyl)-(C2-alkyl)- , or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.
In accordance with a fourth aspect, the present invention covers compounds of general formula (I ), supra, in which :
X1 represents NR3, X2 represents CR6,
R' represents -N(R10)R11,
R2 represents hydrogen,
R3 represents hydrogen,
R4 represents hydrogen,
R5 represents hydrogen,
R6 represents hydrogen,
R7 represents hydrogen,
R8 represents a group selected from
aryl, aryl-(Ci-C2-alkyl)-,
wherein aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
fluorine and chlorine,
R10 and R" are independently of each other selected from :
hydrogen, C,-C alkyl, (C,-alkyl)2N(C2-alkyl)-, (piperidin-1-yl)-(C2-alkyl)-,
or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.
In accordance with a fifth aspect, the present invention covers a compound of general formula (I), supra, which is selected from the group consisting of :
N5-{4-[(3-chlorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin-1-yl)ethyl]-1H-imidazole-4,5- dicarboxamide,
N5-{4-[(3-chlorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin-1-yl)ethyl]-1H-imidazole-4,5- dicarboxamide formic acid salt,
N5-{4-[(2-chloro-4-fluorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin-1-yl)ethyl]-1H- imidazole-4,5-dicarboxamide,
N5-{4-[(2-chloro-4-fluorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin-1-yl)ethyl]-1H- imidazole-4,5-dicarboxamide formic acid salt,
N5-{4-[(3,5-dichlorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin-1-yl)ethyl]-1 H-imidazole- 4,5-dicarboxamide,
N5-{4-[(3,5-dichlorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole- 4,5-dicarboxamide formic acid salt,
N -{4-[(3,5-dichlorophenyl)carbamoyl]phenyl}-N5-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole- 4,5-dicarboxamide,
N4-{4-[(3,5-dichlorophenyl)carbamoyl]phenyl}-N5-methyl-1 H-imidazole-4,5- dicarboxamide,
N4-{4-[(2 -dichlorophenyl)carbamoyl]phenyl}-N5-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole- 4,5-dicarboxamide,
N4-{4-[(3-chlorophenyl)carbamoyl]phenyl}-N5-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4,5- dicarboxamide,
N4-{4-[(3-chlorophenyl)carbamoyl]phenyl}-N5-methyl-1 H-imidazole-4,5-dicarboxamide,
N4-{4-[(2,3-dichlorophenyl)carbamoyl]phenyl}-N5-methyl-1 H-imidazole-4,5- dicarboxamide,
N4-(4-{[1 -(2-chlorophenyl)ethyl]carbamoyl}phenyl)-N5-[2-(piperidin-1 -yl)ethyl]-1 H- imidazole-4,5-dicarboxamide,
N4-(4-{[1 -(2-chlorophenyl)ethyl]carbamoyl}phenyl)-N5-methyl-1 H-imidazole-4,5- dicarboxamide,
N4-{4-[(2,3-dichlorobenzyl)carbamoyl]phenyl}-N5-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole- 4,5-dicarboxamide,
N4-{4-[(2,3-dichlorobenzyl)carbamoyl]phenyl}-N5-methyl-1 H-imidazole-4,5- dicarboxamide,
N5-{4-[(2-chlorophenyl)carbamoyl]phenyl}-N4-methyl-1 H-imidazole-4.5-dicarboxamide,
N5-{4-[(2-chloro-4-fluorophenyl)carbamoyl]phenyl}-N -methyl-1 H-imidazole-4,5- dicarboxamide,
N5-{4-[(4-fluorophenyl)carbamoyl]phenyl}-N4-methyl-1 H-imidazole-4,5-dicarboxamide,
N4- [2- (di methylamino)ethyl] - N5-{4- [(4-fluorophenyl)carbamoyl] phenyl}- 1 H-imidazole- 4,5-dicarboxamide,
N5-{4-[(4-fluorophenyl)carbamoyl]phenyl}-N4-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4,5- dicarboxamide, N5-{4-[(2-chloro-4-fluorophenyl)carbamoyl]phenyl}-N -[2-(piperidin-1-yl)ethyl]-1H- imidazole-4,5-dicarboxamide,
N5-{4-[(2^hloro-4-fluorophenyl)carbamoyl]phenyl}-N4 2-(dimethylamino)ethyl]-1H- imidazole-4,5-dicarboxamide,
N5-{4-[(2 h[orophenyi)carbamoy[]pheny[}-N4-[2-(p^eridin-1-yl)ethyi]-1H-imidazole-4,5- dicarboxamide, and
N5-{4-[(2 hlorophenyl)carbamoyl]phenyl}-N4-[2-(dimethylamino)ethyl]-1H-imidazole-4,5- dicarboxamide,
or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
X1 represents NR3,
X2 represents CR6,
R1 represents -N(R!0)R",
R2 represents hydrogen,
R3 represents hydrogen,
R4 represents hydrogen,
R5 represents hydrogen,
R6 represents hydrogen,
R7 represents hydrogen,
R8 represents a group selected from
aryl, aryl-(Ci-alkyl)-,
wherein aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
halogen,
R'° and R11 are independently of each other selected from :
hydrogen, (Ci-alkyl)i.N(Cralkyl)-, (6-membered heterocycloalkyl)-(C?-alkyl)-, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
X1 represents NR\
X2 represents CR6,
R1 represents -N (R, 0)R" ,
R2 represents hydrogen ,
R3 represents hydrogen ,
R4 represents hydrogen ,
R5 represents hydrogen ,
R6 represents hydrogen ,
R7 represents hydrogen ,
R8 represents a group selected from
aryl, aryl-(Ci -alkyl)- ,
wherein aryl is optionally substituted with one, or two substituents, which are independently of each other selected from :
fluorine and chlorine,
R'° and R" are independently of each other selected from :
hydrogen , (C, -alkyl)2N(C2-alkyl)- , (piperidin-1 -yl)-(C2-alkyl)- ,
or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof , or a mixture of same.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
X1 represents NR3 or 0. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
X1 represents NR3.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
X1 represents 0.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
X2 represents CR6 or N ,
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
X2 represents CR6.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
X2 represents N.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R1 represents a group selected from :
-OR9, and -N(R,0)R" .
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R1 represents a group selected from :
-OR9. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R1 represents a group selected from :
-N(R,0)R" .
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R2 represents a group selected from :
hydrogen, G-Cralkyl, and CrCt-cycloalkyl.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R2 represents a group selected from :
hydrogen, and G-a ky .
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R2 represents hydrogen.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R5 represents a group selected from :
hydrogen, and Ci -Cs-alkyl.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R5 represents a group selected from :
hydrogen, and G-alkyl. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R5 represents hydrogen.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R6 represents a group selected from :
hydrogen, and halogen.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R6 represents a group selected from :
hydrogen, and fluorine.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R6 represents hydrogen. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R8 represents a group selected from :
aryl, aryl-(G-G-alkyl)-, heteroaryl, and heteroaryl- (G-G-alky )-,
wherein aryl and heteroaryl groups are optionally substituted with one, two or three substituents, which are independently of each other selected from :
G-G-alkyl, G-G-alkoxy, G-G-hydroxyalkyl, G-G-cycloalkyl, G-G-cycloalkoxy, G-G-haloalkyl, G-Grhaloalkoxy, halogen, cyano, nitro, hydroxy, -N(R'°)R", R'°(R")N-(G-G-alkyl)-, R'°(R")N-(G-G-alkoxy)-. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R8 represents a group selected from :
aryl, aryl-(Ci-Ct-alkyl)-, heteroaryl, and heteroaryl- (G-Oalkyl)-,
wherein aryl and heteroaryl groups are optionally substituted with one, two or three substituents, which are independently of each other selected from :
CrG-alkyl, G-Cn-alkoxy, G-Crhydroxyalkyl, G-C^-haloalkyl, halogen, cyano, -N(R,0)R".
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R8 represents a group selected from :
aryl, aryl-(G-C2-alkyl)-,
wherein aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
halogen.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R8 represents a group selected from :
aryl, aryl-(G-C2-alkyl)-,
wherein aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
fluorine and chlorine.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R8 represents aryl optionally substituted with one, two or three substituents, which are independently of each other selected from :
fluorine and chlorine. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R8 represents aryl-(Ci -Cralkyl)- wherein aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
fluorine and chlorine.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R8 represents a group selected from :
aryl, aryl-(d-Cralkyl)-,
wherein aryl is optionally substituted with one, or two substituents, which are independently of each other selected from :
fluorine and chlorine.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R8 aryl, wherein aryl is optionally substituted with one, or two substituents, which are independently of each other selected from :
fluorine and chlorine.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R8 represents aryl-(Ci -G-alkyl)-, wherein aryl is optionally substituted with one, or two substituents, which are independently of each other selected from :
fluorine and chlorine.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R9 represents Ci-Cj-alkyl. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R9 represents Ci -C2-alkyl. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R'° and R11 are independently of each other selected from :
hydrogen, Ci -Ca-alkyl, C3-C4-cycloalkyl, (C3-C4-cycloalkyl)-(0-C4-alkyl)-,
CrC4-hydroxyalkyl, (G -C3-alkoxy)-(C2-C4-alkyl)-, Ci -haloalkyl, H2N-(C2-C4-alkyl)-, (Ci-C3-alkyl)N{H)(C2-C4-alkyl)-, (C, -C3-alkyl)2N(C2-C4-alkyl)-, 4-6 membered heterocycloalkyl, (4- to 6-membered heterocycloalkyl)- (C2-C4-alkyl)-, wherein 4- to 6-membered heterocycloalkyl groups are optionally substituted with one or two substituents, which are independently of each other selected from :
Ci-alkyl, d-haloalkyl, Ci -alkoxy, Ci -hydroxyalkyl, Ci-haloalkoxy, halogen, and hydroxy; or,
R'° and R" together with the nitrogen atom to which they are attached form a 4- to 6-membered heterocycloalkyl group, in which one carbon atom is optionally replaced by a further heteroatom-containing group selected from NR12, and 0, in which heterocycloalkyl group one additional ring atom is optionally replaced by C(=0), said 4-to 6-membered heterocycloalkyl group being optionally substituted with one or two substituents, which are independently of each other selected from :
Ci-alkyl, O-haloalkyl, Ci -alkoxy, Ci -haloalkoxy, halogen, and hydroxyl. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R'° and R11 are independently of each other selected from :
hydrogen, G -G,-alkyl, C3-C4-cycloalkyl, (C3-C4-cycloalkyl)-(G -C4-alkyl)-,
C2-C4-hydroxyalkyl, (G -C3-alkoxy)-(C2-C4-alkyl)-, CrC4-haloalkyl, H2N-(C2-C4-alkyl)-, (Ci-C3-alkyl)N(H)(C2-C4-alkyl)-, (C, -Cralkyl)2N(C2-C4-alkyl)-, 4-6 membered heterocycloalkyl, (4- to 6-membered heterocycloalkyl)- (C2-C4-alkyl)-, wherein 4- to 6-membered heterocycloalkyl groups are optionally substituted with one or two substituents, which are independently of each other selected from :
G-alkyl, G-haloalkyl, G -a koxy, G -hydroxyalkyl, G-haloalkoxy, halogen, and hydroxy I.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R'° is selected from hydrogen, and G -Ct-alkyl, preferably hydrogen,
and
R" is selected from :
hydrogen, G -Ct-alkyl, G3-G.-cycloalkyl, (G-G-cycloalkyl)-(G-G-alkyl)- ,
GrG-hydroxyalkyl, (G-G-alkoxy)-(G-G-alkyl)-, G-G-haloalkyl,
HzN-(G-G-alkyl)-, (G-G-alkyl)N(H)(G-G-alkyl)-, (G -G-alkyl)2N(GrG-alkyl)-, 4-6 membered heterocycloalkyl, (4- to 6-membered heterocycloalkyl)- (G-G-alkyl)-,
wherein 4- to 6-membered heterocycloalkyl groups are optionally substituted with one or two substituents, which are independently of each other selected from :
G-alkyl, G-haloalkyl, G -alkoxy, G -hydroxyalkyl, G-haloalkoxy, halogen, and hydroxyl.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R'° and Rn together with the nitrogen atom to which they are attached form a 4- to 6-membered heterocycloalkyl group, in which one carbon atom is optionally replaced by a further heteroatom-containing group selected from NR'2, and 0, in which heterocycloalkyl group one additional ring atom is optionally replaced by C(=0),
said 4-to 6-membered heterocycloalkyl group being optionally substituted with one or two substituents, which are independently of each other selected from :
G-alkyl, G-haloalkyl, G -alkoxy, G -haloalkoxy, halogen, and hydroxyl. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R'° and R" are independently of each other selected from :
hydrogen, G-G-alkyl, Cr -cycloalkyl, (CrC4-cycloalkyl)-(Ci -C3-alkyl)-, C?-C3-hydroxyalkyl, (Cralkoxy)- (C2-C)-alkyl)-, G-C3-haloalkyl, H2N-(C2-C3-alkyl)-, (G- alkyl)N(H)(C7-C3-alkyl)-, (Ci-alkyl)2N(C2-C3-alkyl)-, (4-6 membered heterocycloalkyl)- (C2-C3- alkyl)-, wherein 4-6-membered heterocycloalkyl groups are optionally substituted with one or two substituents, which are independently of each other selected from :
G-alkyl, G-haloalkyl, Ci-alkoxy, G-hydroxyalkyl, G-haloalkoxy, halogen, and hydroxy.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R'° is selected from hydrogen, and Ci -Ca-alkyl, preferably hydrogen,
and
R" is selected from :
hydrogen, Ci -Ca-alkyl, C3-Grcycloalkyl, (C3-G,-cycloalkyl)-(G -G-alkyl)-,
C?-C3-hydroxyalkyl, (G-alkoxyHG-G-alkyl)-, Ci- rhaloalkyl, H2N-(C2-G-alkyl)-, (G- alkyl)N(H)(C2-C3-alkyl)-, (Ci-alkyl)2N(C2-C3-alkyl)-, and (4-6 membered heterocycloalkyl)- (C2-C3-alkyl)-, wherein 4-6-membered heterocycloalkyl groups are optionally substituted with one or two substituents, which are independently of each other selected from :
Ci-alkyl, Ci-haloalkyl, Ci-alkoxy, G-hydroxyalkyl, G-haloalkoxy, halogen, and hydroxy.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R'° and R11 are independently of each other selected from : hydrogen, Ci-Cj-alkyl, CrC4-cycloalkyl, (CrC4-cycloalkyl)-(G-alkyl)-,
CrCrhydroxyalkyl, (G-alkoxy)-(C2-Cralkyl)-, G-Crhaloalkyl, H2N-(C?-alkyl)-, (G- alkyl)N(H)(G-alkyl)-, (C,-alkyl N(C2-alkyl)-, (6-membered heterocycloalkyl)-(Gralkyl)-.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R10 is selected from hydrogen, and G-G-alkyl, preferably hydrogen,
and
R" is selected from :
hydrogen, G-Gi-alkyl, G-G-cycloalkyl, (GrG-cycloalkyl)-(G-alkyl)-,
G-G-hydroxyalkyl, (G-alkoxy)- (G-G-alkyl)-, G-Grhaloalkyl, H2N-(Cralkyl)-, (G- alkyl)N(H)(G-alkyl)-, (Ci-alkyl)2N(C2-alkyl)-, (6-membered heterocycloalkyl)-(C2-alkyl)-.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R10 and R" are independently of each other selected from :
hydrogen, C C2-alkyl, (Cralkyl)2N(C2-alkyl)-, (piperidin-1-yl)-(C2-alkyl)-.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R'° is selected from hydrogen, and CrC2-alkyl, preferably hydrogen,
and
R11 is selected from :
hydrogen, G-C2-alkyl, (Ci-alkyl)2N(C2-alkyl)-, and (piperidin-1-yl)-(C2-alkyl)-.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R'° and R" are independently of each other selected from :
hydrogen, (Ci-alkyl)2N(C2-alkyl)-, (6-membered heterocycloalkyl)-(C2-alkyl)-. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R'° and R" are independently of each other selected from :
hydrogen, (C,-alkyl)2N(Cralkyl)-, (piperidin-1 -yl)-(C2-alkyl)-.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R10 is selected from hydrogen, and Ci-Cralkyl, preferably hydrogen,
and
Rn is selected from :
(Ci-alkyl)2N(C2-alkyl)-, and (6-membered heterocycloalkyl)-(C?-alkyl)-.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R'° is selected from hydrogen, and Ci-C?-alkyl, preferably hydrogen,
and
R" is selected from :
(C,-alkyl)2N(C2-alkyl)-, and (piperidin- 1 -yl)-(C2-alkyl)-. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R'2 represents a group selected from :
hydrogen, and G-alkyl. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R12 represents a group selected from :
hydrogen. In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), wherein :
R12 represents a group selected from :
C-alkyl.
In a further embodiment of the above-mentioned aspects, the invention relates to compounds of formula (I), according to any of the above-mentioned embodiments, in the form of or a stereoisomer, a tautomer, an N- oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.
It is to be understood that the present invention relates to any sub-combination within any embodiment or aspect of the present invention of compounds of general formula (I), supra.
More particularly still, the present invention covers compounds of general formula (I) which are disclosed in the Example section of this text, infra.
In accordance with another aspect, the present invention covers methods of preparing compounds of the present invention, said methods comprising the steps as described in the Experimental Section herein.
Particularly, another aspect of the present invention relates to a method of preparing a compound of general formula (I) as defined herein, said method comprising the step of allowing an intermediate compound of general formula (III) :
Figure imgf000039_0001
(Hi) in which Xi represents N, and R1 , and R2 are as defined herein for the compound of general formula (I),
to react with a compound of general formula (II) :
Figure imgf000040_0001
in which X2, R , R5, R7, and R8 are as defined herein for the compound of general formula (I), thereby giving a compound of general formula (I) :
Figure imgf000040_0002
(I) in which X, represents NR3, and X¾, R! , R2, R3, R5, R7, and R8 are as defined herein for the compound of general formula (I).
Another aspect of the present invention relates to a method of preparing a compound of general formula (I) as defined herein, said method comprising the step of allowing an intermediate compound of general formula (3-4) :
Figure imgf000040_0003
in which Xi represents NR3, X2, R1 , R2, R3, R4, R5, and R9 are as defined herein for the compound of general formula (I) and R13 represents R9, or H. to react with a compound of general formula (1 -2)
,8
HN
I
1-2
in which R7, and R8 are as defined herein for the compound of general formula (I),
thereby giving a compound of general formula (I) :
Figure imgf000041_0001
in which Xi represents NR3, and X?, R\ R2, R4, R5, R7, and R8 are as defined herein for the compound of general formula (I). Another aspect of the present invention relates to a method of preparing a compound of general formula (I ) as defined, said method comprising the step of allowing an intermediate compound of general formula (VI) :
Figure imgf000042_0001
in which Xi represents NR3, X2, R2, R3, R4, R5, R7 and R8 are as defined herein for the compound of general formula (I),
to react with a compound of general formula (1 -2) :
R1-H
3-2 in which R1 represents represents N(R10)R11 and R'°, and R11 are as defined herein for the compound of general formula (I), thereby giving a compound of general formula (I) :
Figure imgf000042_0002
(I) in which X1 represents NR3, R1 represents N(R'°)R" and X2, R2, R3, R4, R5, R7, R8, R'°, and R" are as defined herein for the compound of general formula (I).
Another aspect of the present invention relates to a method of preparing a compound of general formula (I) as defined herein, said method comprising the step of allowing an intermediate compound of general formula (IV) :
Figure imgf000043_0001
in which X1 represents N , and X2, R2, R4, R5, R7 and R8 are as defined herein for the compound of general formula (I),
to react with a compound of general formula (1 -2) :
R1-H
3-2
in which R1 is as defined herein for the compound of general formula (I ),
thereby giving a compound of general formula (I )
Figure imgf000043_0002
(I)
ί
in which X1 represents NR3 and X2, R\ R2, R3, R4, R5, R7, and R8 are as defined herein for the compound of general formula (I ).
Another aspect of the present invention relates to a method of preparing a compound of general formula (I ) as defined herein , said method comprising the step of allowing an intermediate compound of general formula (VII ) :
Figure imgf000044_0001
in which X2, R4, R5, R7, and R8 are as defined herein for the compound of general formula (I), to react with a compound of general formula (4-1 ) :
Figure imgf000044_0002
4-1
in which R! , and R2 are as defined herein for the compound of general formula (I),
thereby giving a compound of general formula (I)
Figure imgf000044_0003
(I) in which X1 represents 0, and, X2, R1 , R2, R4, R5, R7, and R8 are as defined herein for the compound of general formula (I). Another aspect of the invention is intermediate (III) or a salt thereof:
Figure imgf000045_0001
in which Xi represents N , R1 , and R2 are as defined herein for the compound of general formula (I ).
Another aspect of the invention is intermediate (3-4) or a salt thereof:
Figure imgf000045_0002
in which Xt represents NR3, X2, R\ R2, R3, R4, R5, and R9 are as defined herein for the compound of general formula (! ) and R13 represents R9, or H.
Another aspect of the invention is intermediate (VI ) or a salt thereof:
Figure imgf000045_0003
in which X, represents NR3, X2, R2, R3, R4, R5, R7 and R8 are as defined herein for the compound of general formula (I ). Another aspect of the invention is intermediate (IV) or a salt thereof:
Figure imgf000046_0001
in which X1 represents N, and X2, R2, R4, R5, R7 and R8 are as defined herein for the compound of general formula (I).
Another aspect of the invention is intermediate (VII) or a salt thereof:
Figure imgf000046_0002
in which X2, R4, R5, R7, and R8 are as defined herein for the compound of general formula
In accordance with a further aspect, the present invention covers the use of the intermediate compounds of general formula (III), (3-4), (VI), (IV) and (VII), or a salt thereof for the preparation of a compound of general formula (I)
Figure imgf000047_0001
(I) or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N- oxide, tautomer or stereoisomer,
in which X1, X2, R1 , R2, R4, R5, R7, and R8 are as defined herein for the compound of general formula (I).
Another aspect of the invention relates to the intermediates or salts thereof described herein and their use for preparing a compound of formula (I) as defined supra or an N- oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
The intermediates used for the synthesis of the compounds of claims 1 to 5 as described below, as well as their use for the synthesis of the compounds of claims 1 to 5, are one further aspect of the present invention. Preferred intermediates are the Intermediate Examples as disclosed below.
EXPERIMENTAL SECTION
1. Syntheses of Compounds (Overview):
The compounds of the present invention can be prepared as descibed in the following section. Schemes 1 to 6 and the procedures described below illustrate general synthetic routes to the compounds of general formula (I ) of the invention and are not intended to be limiting. It is clear to the person skilled in the art that the order of transformations as exemplified in Schemes 1 to 6 can be modified in various ways. The order of transformations as exemplified in the Schemes 1 to 6 are therefore not intended to be limiting, in addition, interconversion of any of the substituents, R1 , R2, R4, R5 , R7 and R8 can be achieved before and/or after the exemplified transformations. These modifications can be such as the introduction of protecting groups, cleavage of protecting groups, exchange, reduction or oxidation of functional groups, halogenation, metallation, substitution or other reactions known to the person skilled in the art. These transformations include those which introduce a functionality which allows for further interconversion of substituents. Appropriate protecting groups and their introduction and cleavage are well-known to the person skilled in the art (see for example T.W. Greene and P.G.M. Wuts in Protective Groups in Organic Synthesis, 3rd edition, Wiley 1999). Specific examples are described in the subsequent paragraphs. Further, it is possible that two or more successive steps may be performed without work-up being performed between said steps, e.g. a "one- pot" reaction, as is well-known to the person skilled in the art.
All reagents used for the preparation of the compounds of the invention are either commercially available or can be prepared as described.
1.1 Synthesis of aromatic amines
Aromatic amines as intermediates for the synthesis of compounds of the invention are either commercially available or can be synthesized as depicted in scheme 1 .
Figure imgf000048_0001
Scheme 1 : Synthesis of aromatic amines, wherein X2, R4, R5, R7 and R8 are as defined for the compound of general formula (I) supra, and in which PG represents a protecting group, such as a BOC group, and W represents a hydroxy group or a chlorine atom.
Starting from an aromatic carboxylic acid of type 1 -1 , wherein the amino function might be bearing a protecting group such as, for example, a BOC group, amides of type 1 -3 can be obtained by reaction with amines of type 1-2 in the presence of a coupling agent such as, for example, HATU or PyBOP, or via the formation of the corresponding acid chloride of type 1 - 1.
Alternatively, starting from an aromatic carboxylic acid of type 1 - 1 , wherein PG-N-R4 represents a nitro group in structures 1 - 1 and 1 -3, amides of type 1 -3 can be obtained by reaction with amines of type 1 -2 in the presence of a coupling agent such as, for example, HATU or PyBOP, or via the formation of the corresponding acid chloride of type 1 - 1.
Deprotection of the protected amine of type 1 -3, in the case of a BOC-protecting group for example employing TFA or hydrochloric acid, or in the case of PG-N-R4 represents a nitro group reduction of the nitro-moiety, for example using zinc powder under acidic conditions, i.e. employing aqueous hydrochloric acid, or a palladium on charcoal catalyst with hydrogen gas, results in compounds of general formula (II), which can then be transformed further as decribed, for example, in schemes 3 to 6. 1.2 Synthesis of imidazole derivatives
One possible synthesis route for the compounds of this invention is depicted in schemes 2 and 3.
Figure imgf000049_0001
2-1 2-2 2-3
Scheme 2: Synthesis of 3,4 dicarboxylic acid substituted imidazoles, wherein X1 represents NR3, and R2 and R3 are as defined for the compounds of general formula (I) supra. Commercially available benzene-1 ,2-diamine 2-1 can be reacted with carboxyclic acids at elevated temperatures to give compounds of type 2-2. Treatment of compound 2-2. for example with hydrogen peroxide under acidic conditions and elevated temperature, yields compounds of type 2-3.
The synthesis of some of the compounds claimed in the present invention employs the dicarboxylic acids of type 2-3 as depicted in scheme 3 to 5.
Figure imgf000050_0001
Scheme 3: Synthesis of compounds starting from dicarboxylic acid precursors, wherein Xt represents NR\ except for compounds 3-1 and (III) wherein X1 represents N, and X2, R' , R2, R\ R4, R5, R7, R8, R9, R'° and R1 1 are as defined for the compound of general formula (I) supra, and R13 represents R9 or H. Starting from the corresponding 1 H-imidazole-4,5-dicarboxylic acid derivative 2-3, after treatment with thionylchloride at elevated temperature 5, 10-dioxo-5H, 10H-diimidazo[1 ,5- a: 1 ', 5'-d]pyrazine-1 ,6-dicarbonyl dichlorides of type 3- 1 are obtained. Compounds of type 3- 1 can be reacted with suitable nucleophiles, such as, for example, amines or alcohols of type 3-2 in presence of a suitable base, for example N,N- diisopropylethylamine, to give compounds of general formula (III).
Compounds of type (III) may serve as starting materials for several transformations:
Compounds of general formula (I) can be obtained directly by reacting compounds of general formula (III) with a fully decorated aromatic amine of general formula (II) at room temperature or at elevated temperature (e.g. under reflux).
Alternatively, an intermediate of type 3-4 can be obtained by reacting a compound of general formula (III) with a suitably substituted aromatic amine of type 3-3 at room temperature or elevated temperature (e.g. under reflux). Esters of type 3 -4a (wherein R13 = R9) can be transformed directely to compounds of general formula (I) by reaction with a suitable nucleophile of type 1 -2 at elevated temperature (e.g. under reflux), optionally in the presence of reagents such as DABAL. Alternatively, compounds of general formula (I) can be obtained in a 2 step procedure from esters of type 3-4a via, first, ester hydrolysis to afford carboxylic acid 3-4b, for example, under basic conditions, followed by standard amide bond forming reactions, for example with amines of type 1 -2 in the presence of a coupling agent such as, for example, T3P, HATU, PyBOP, or alternatively in a three step procedure after hydrolysis of the ester, generation of corresponding acid chloride, for example using thionylchloride or 1 -chloro- N, N,2-tri methyl- 1 -propenylamine and reaction with amines of type 1 -2 under basic conditions in presence of , for example, N, N-diisopropylethylamine or pyridine.
Esters of general formula (I) (in which R1 = OR9) can be transformed into amides of general formula (I) (in which R1 = N(R'°)Rn ), according to the invention, for example by treatment with different amines of formula HN(R'°)R" , optionally in presence of a base, such as, for example, N,N-diisopropylethylamine, or alternatively in a two step procedure consisting of ester hydrolysis, for example using sodium hydroxide followed by standard amide bond formation in presence of amines and coupling agents such as HATU or alternatively in a three step procedure after hydrolysis of the ester, generation of corresponding acid chloride, for example using thionylchloride and reaction with amines under basic conditions in presence of. for example. N, N-diisopropylethylamine.
An alternative synthesis route of imidazole derivatives of the present invention is depicted in scheme 4.
Figure imgf000052_0001
(I)
Scheme 4: Alternative Synthesis of imidazole derivatives, wherein X1 represents NR (except for compounds 3-1 and (IV) wherein X1 represents N) and X2, R' , R2, R3, R4, R5, R7 and R8 are as defined for the compound of general formula (I) supra. Starting from compounds of type 3-1 , upon reaction with aromatic amines of general formula (II) in the presence of a base such as, for example N ,N-diisopropylethylamine, compounds of general formula (IV) can be obtained.
Compounds of general formula (IV) can be transformed to compounds of general formula (I) by reaction with nucleophiles of type 3-2, such as, for example, amines HN(R'°)R" , or alcohols HOR9, optionally in the presence of a base, such as, for example N, N- diisopropylethylamine.
Figure imgf000053_0001
Scheme 5: Alternative Synthesis of imidazole derivatives starting from dicarboxylic acid precursors, wherein X1 represents NR3 (except for compounds 3-1 and (V) wherein X1 represents N) and X2, R2, R3, R4, R5, R7, R8, R9, R10 and R" are as defined for the compound of general formula (I) supra and R1 represents N(R'°)R" . Compounds of type 3- 1 can be reacted with suitable nucleophiles, such as, for example, phenol (5- 1 ) in presence of a suitable base, for example N ,N-diisopropylethylamine, to give compounds of general formula (V).
Compounds of general formula (V) can react with a fully decorated aromatic amine of general formula (II) at room temperature or elevated temperature (e.g. under reflux) to give compounds of the general formula (VI).
Compounds of general formula (VI) can be transformed into amides of general formula (I), according to the invention, for example by treatment with different amines of type HN(R,0)(R, , )) optionally in presence of a base, such as, for example, N,N- diisopropylethylamine.
1.3 Synthesis of oxazole derivatives
Yet another possible synthesis route for the compounds of this invention is depicted in scheme 6.
Figure imgf000054_0001
C R1 = OR9
R1 = N(R10)R11
Scheme 6: Synthesis of oxazole derivatives of the present invention, wherein X1 represents 0, and, X7, R\ R2, R4, R5, R7, R8, R9, R'° and R" as defined for the compound of general formula (I) supra. Compounds of type (II) can be transformed into compunds of type (VII) by reaction with oxalyl chloride.
Reaction of compounds of general formula (VII) with alkyl isocyanoacetates of type 4-1 in presence of, for example, imidazole and triethylamine yields esters of general formula (I) (where R1 = OR9) as claimed in this invention.
Esters of general formula (I) (in which R1 = OR9) can be transformed into amides of general formula (I) (in which R1 = N(R'°)R" ), according to the invention, for example by treatment with different amines, optionally in presence of a base, such as, for example, N,N- diisopropylethylamine, or alternatively in a two step procedure consisting of ester hydrolysis, for example using sodium hydroxide, followed by standard amide bond formation in presence of amines and coupling agents such as HATU, or alternatively in a three step procedure, after hydrolysis of the ester, generation of the corresponding acid chloride, for example using thionylchloride, and reaction with amines under basic conditions in presence of, for example, N,N-diisopropylethylamine.
General part
Chemical names were generated using ACD/Name Batch Version 12.02.. In case there is discrepancy between the chemical name of a compound and its chemical structure, the chemical structure shall prevail. In some cases generally accepted names of commercially available reagents were used in place of ACD generated names.
The following table lists the abbreviations used in this paragraph and in the Intermediates and Examples section as far as they are not explained within the text body. A comprehensive list of the abbreviations utilized by organic chemists of ordinary skill in the art appears presented in the first issue of each volume of the Journal of Organic Chemistry; this list is typically presented in a table entitled Standard List of Abbreviations. The abbreviations contained therein, and all abbreviations utilized by organic chemists of ordinary skill in the art are hereby incorporated by reference.
Figure imgf000055_0001
d doublet (NMR)
dd doublet of doublet (NMR)
dt doublet of triplet (NMR)
DMSO dimethylsulfoxide
EDTA ethylenediaminetetraacetic acid
ESI electrospray (ES) ionisation
h hour(s)
1 - [bis(dimethylamino)methylene] - 1 H- 1 ,2, 3-triazolo[4, 5-b] -pyridinium
HATU
3-oxid hexafluorophosphate
HQ hydrochloric acid
HPLC high performance liquid chromatography
HRP horseradish peroxidase
LC-MS liquid chromatography mass spectrometry
m multiplet (NMR)
min minute(s)
MS mass spectrometry
MTP microtiter plate
nuclear magnetic resonance spectroscopy : chemical shifts (δ) are
NMR given in ppm. The chemical shifts were corrected by setting the
DMSO signal to 2.50 ppm using unless otherwise stated.
NAD' nicotinamide adenine dinucleotide
PBS phosphate buffered saline
PG protecting group
Ph phenyl
PyBOP benzotriazol-1 -yl-oxytripyrrolidinophosphonium hexafluorophosphate q quartet (NMR)
quin quintet (NMR) s singulet (NMR)
SPA Scintillation proximity assay
T3P 1 -propanephosphonic anhydride
TBAF tetrabutylammonium fluoride
t triplet (NMR)
td triplet of doublet (NMR)
TFA trifluoro acetic acid
THF tetrahydrofuran
[:,H]- tritium
δ chemical shift
Other abbreviations have their meanings customary per se to the skilled person.
The various aspects of the invention described in this application are illustrated by the following examples which are not meant to limit the invention in any way. NMR data:
NMR peak forms are stated as they appear in the spectra, possible higher order effects have not been considered.
The 1 H-NMR data of selected examples are listed in the form of Ή-NMR peaklists. For each signal peak the δ value in ppm is given, followed by the signal intensity, reported in round brackets. The δ value-signal intensity pairs from different peaks are separated by commas. Therefore, a peaklist is described by the general form: δι (intensity! ), δ? (intensity?), ... , δ,- (intensity.), ... , δπ (intensity,,).
The intensity of a sharp signal correlates with the height (in cm) of the signal in a printed NMR spectrum. When compared with other signals, this data can be correlated to the real ratios of the signal intensities. In the case of broad signals, more than one peak, or the center of the signal along with their relative intensity, compared to the most intense signal displayed in the spectrum, are shown. A Ή-NMR peaklist is similar to a classical ' H-NMR readout, and thus usually contains all the peaks listed in a classical NMR interpretation. Moreover, similar to classical 'H-NMR printouts, peaklists can show solvent signals, signals derived from stereoisomers of target compounds (also the subject of the invention), and/or peaks of impurities. The peaks of stereoisomers, and/or peaks of impurities are typically displayed with a lower intensity compared to the peaks of the target compounds (e.g. , with a purity of >90%). Such stereoisomers and/or impurities may be typical for the particular manufacturing process, and therefore their peaks may help to identify the reproduction of our manufacturing process on the basis of "by-product fingerprints". An expert who calculates the peaks of the target compounds by known methods (MestReC, ACD simulation , or by use of empirically evaluated expectation values) , can isolate the peaks of target compounds as required , optionally using additional intensity filters. Such an operation would be similar to peak-picking in classical Ή-NMR interpretation. A detailed description of the reporting of NMR data in the form of peaklists can be found in the publication "Citation of NMR Peaklist Data within Patent Applications" (cf. Research Disclosure Database Number 605005, 2014, 01 Aug 2014, or http: / /www. researchdisclosure.com/searching-disclosures). In the peak picking routine, as described in the Research Disclosure Database Number 605005, the parameter "MinimumHeight" can be adj usted between 1 % and 4%. Depending on the chemical structure and/or depending on the concentration of the measured compound it may be reasonable to set the parameter "MinimumHeight" < 1 %.
Analytical HPLC Methods:
Method 1
Instrument: Waters Acquity UPLCMS SingleQuad; Column: Acquity UPLC BEH C18 1 .7 μνη, 50x2.1 mm; eluent A: water + 0.1 vol % formic acid (99%), eluent B: acetonitrile; gradient: 0- 1 .6 min 1 -99% B, 1 .6-2.0 min 99% B; flow 0.8 ml/ min; temperature: 60 C; DAD scan: 210-400 nm.
Method 2lnstrument: Waters Acquity UPLCMS SingleQuad; Column: Acquity UPLC BEH C18 1 .7 μιτι , 50x2.1 mm; eluent A: water + 0.2 vol % aqueous ammonia (32%), eluent B: acetonitrile; gradient: 0-1 .6 min 1 -99% B, 1 .6-2.0 min 99% B; flow 0.8 ml/ min; temperature: 60 C; DAD scan: 210-400 nm.
Method 3:
Instrument: Waters Acquity UPLCMS SingleQuad; Column: Acquity UPLC BEH C18 1 .7 50x2.1 mm; eluent A: water + 0.1 vol % formic acid (99%), eluent B: acetonitrile; gradient: 0- 1 .6 min 1 -99% B, 1 .6-2.0 min 99% B; flow 0.8 ml/ min; temperature: 60 C; DAD scan: 210-400 nm.
Method 4: Instrument: Waters Acquity UPLCMS Tof; column: Kinetex C 18 (Phenomenex) 2.6 μιτι, 50x2.1 mm; eluent A: water + 0.05 Vol-% formic acid (99%), eluent B: acetonitrile + 0.05% formic acid; gradient: 0-0.2 min 98% A, 0.2-1 .7 min 98-10% A, 1 .7-1.9 min 10% A, 1.9-2.0 min 10-98% A, 2.0-2.5 min 98% A; flow 1.3 ml/min; temperature: 60 C; DAD scan: 210-400 nm
Method 5:
Instrument: Agilent 1290 UHPLCMS Tof; column: BEH C 18 (Waters) 1 .7 μιτι, 50x2.1 mm; eluent A: water + 0.05 Vol-% formic acid (99%), eluent B: acetonitrile + 0.05% formic acid; gradient: 0-1 .7 min 98-10% A, 1 .7-2.0 min 10% A, 2.0-2.5 min 10-98% A, flow 1 .2 ml/min; temperature: 60 C; DAD scan: 210-400 nm
Preparative HPLC methods:
Method 6:
Instrument: Waters Autopurification MS SingleQuad; Column: Waters XBrigde C18 5μ 100x30mm; eluent A: water + 0.1 vol % formic acid (99%), eluent B: acetonitrile; gradient eluent A/ eluent B, flow 70 ml/min; temperature: 25 C; DAD scan: 210-400 nm.
Method 7:
Instrument: Waters Autopurification MS SingleQuad; Colum: Waters XBrigde C18 5μ 100x30mm; eluent A: water + 0.2 vol % aqueous ammonia (32%), eluent B: acetonitrile; gradient: eluent A / eluent B; flow 70 ml/min; temperature: 25 C; DAD scan: 210-400 nm.
Intermediates
Intermediate 11
5, 10-dioxo-5H, 1 OH-diimidazo[ 1 , 5-σ: 1 ', 5'-d]pyrazine- 1 , 6-dicarbonyl dich loride
Figure imgf000060_0001
Thionyl chloride (94 ml, 1.29 mol) was added to a suspension of 1H-imidazole-4,5- dicarboxylic acid (25 g, 157 mmol) in toluene (334 ml) and N,N-dimethylformamide (12.1 ml) and the mixture was stirred for 24 h at 80 C. The mixture was concentrated under reduced pressure, 100 ml toluene were added and the mixture was concentrated under reduced pressure to give 35.5 g of the title compound as crude material, which was used at the same day without further purification for subsequent steps.
Intermediate 12
N,N"-dimethyl-5, 10-dioxo-5H, 10H-diimidazo[ 1 , 5-a: 1 ", 5'-d]pyrazine- 1 ,6-dicarboxamide
Figure imgf000060_0002
To 35 g (crude product) of 5, 10-dioxo-5H, 10H-diimidazo[1 ,5-a:1 ',5'-d]pyrazine-1 ,6- dicarbonyl dichloride in 460 mL tetrahydrofuran were added dropwise 153 mL (307 mmol) of a 2M solution of methyl amine in tetrahydrofruan and 68 mL (391 mmol) N,N- diisopropylethylamine. The resulting mixture was stirred for 20 h at room temperature. The precipitate was filtered off and washed with dichloromethame. The obtained solid material was triturated in methanol and dried in vacuum to give 15.2 g of the tiltle compound as a crude product which was used without further purification in the subsequent steps.
Intermediate 13 methyl 4-{[(4-{[2-(piperidin- 1 -yl)ethyl]carbamoyl}-1 H-imidazol-5- yl)carbonyl]amino}benzoate
Figure imgf000061_0001
2-(Piperidin-1 -yl)ethanamine (1 .4 ml, 9.6 mmol) and N, N-diisopropylethylamine (1 .0 ml, 5.7 mmol) were added at 0 C to a suspension of 5, 10-dioxo-5H, 10H-diimidazo[1 ,5-a: 1 ',5'- d]pyrazine-1 ,6-dicarbonyl dichloride (1 .50 g, 4.79 mmol) in tetrahydrofuran (25 ml) and the mixture was stirred over night at room temperature to give 5, 10-dioxo-N, N'-bis[2- (piperidin- 1 -yl)ethyl]-5H, 10H-diimidazo[1 ,5-a: 1 ',5'-d]pyrazine-1 ,6-dicarboxamide.
Tetrahydrofuran (25 ml), methyl 4-aminobenzoate (1 .45 g, 9.58 mmol) and N,N- diisopropylethylamine (8.3 ml, 48 mmol) were added to the above reaction mixture and the mixture was stirred for 3 days at room temperature. For work-up, the solid material was filtrated off, washed with water and tetrahydrofuran and the filtrate was concentrated. The residue was stirred with methanol and the precipitate was collected by filtration and dried under high vacuum at 50 C to give the title compound (960 mg, 70% purity by LC-MS, 35% yield).
LC-MS (Method 1 ): Rt = 0.79 min; MS (ESIpos): m/z = 400.3 [M+H] + Intermediate I4
Lithium 4-{[(4-{[2-(piperidin- 1 -yl)ethyl]carbamoyl}- 1 H-imidazol-5- yl)carbonyl]amino}benzoate
Figure imgf000061_0002
A mixture of methyl 4-{[(4-{[2-(piperidin-1 -yl)ethyl]carbamoyl}-1 H-imidazol-5- yl)carbonyl]amino}benzoate (952 mg, 2.38 mmol), lithium hydroxide (4.8 ml, 1 .0 M aqueous solution, 4.8 mmol), methanol (9.9 ml) and tetrahydrofuran (9.9 ml) was stirred over night at room temperature. For work-up, the reaction mixture was concentrated and then codestilled with toluene (2x) to give the title compound as crude material which used in the next steps without further purification (1 .13 g, 120% yield).
LC-MS (Method 2): R. = 0.53 min; MS (ESIneg): m/z = 384.2 [M-Li] Intermediate I5
4-({[4-(methylcarbamoyl)- 1 H-imidazol-5-yl]carbonyi}amino)benzoic acid
Figure imgf000062_0001
4-aminobenzoic acid (458 mg, purity, 3.31 mmol) and trimethylamine (460 μΐ, 3.3 mmol) were added to a suspension of N,N'-dimethyl-5, 10-dioxo-5H, 10H-diimidazo[1 , 5-a: 1 ',5'- d]pyrazine-1 ,6-dicarboxamide (500 mg, 1.65 mmol) in dichlormethan (12 ml) and the mixture was stirred for 4 hours at room temperature. For work-up, the reaction mixture was concentrated in vacuo and the residue was trituated with dichioromethane to give the title compound as crude material which was used in the next steps without further purification (245 mg, 51 % yield).
LC-MS (Method 1 ): Rt = 0.75 min; MS (ESIpos) m/z = 289.1 [M+H]\ Intermediate 16
tert-butyl {4-[(2-chloro-4-fluorophenyl)carbamoyl]phenyl}carbamate
Figure imgf000062_0002
To a mixture of 4- [(tert-butoxycarbonyl)amino] benzoic acid (5.00 g, 20.4 mmol), N, N- diisopropylethylamine (1 1 ml, 65 mmol) and 1 -[bis(dimethylamino)methylene]-1 H-1 ,2, 3- triazolo[4, 5-b]-pyridinium 3-oxid hexafluorophosphate (1 1 .7 g, 30.7 mmol) in 60 mL N,N- dimethylformamide was added 2-chloro-4-fluoroaniline (3.7 ml, 31 mmol). The mixture was stirred for 3 days at 100 C. For work-up, the reaction mixture was concentrated in vacuo and the residue was trituated with methanol to give the title compound as crude material which was used in the next steps without further purification (3.4 g, 46% yield). LC-MS (Method 1 ): Rt = 1.33 min; MS (ESIpos) m/z = 365.1 [ +H]*.
Intermediate I7
tert-butyl {4-[(4-fluorophenyl)carbamoyl]phenyl}carbamate
Figure imgf000063_0001
To a mixture of 4- [(tert-butoxycarbonyl)amino] enzoic acid (5.00 g, 20.4 mmol), N,N- diisopropylethylamine (11 ml, 65 mmol) and 1 -[bis(dimethylamino)methylene]-1 H-1 ,2,3- triazolo[4,5-b]-pyridinium 3-oxid hexafluorophosphate (11.7 g, 30.7 mmol) in 65 mL N,N- dimethylformamide was added 4-fluoroaniline (2.9 ml, 31 mmol). The mixture was stirred for 1 day at 100 C. For work-up, the reaction mixture was concentrated in vacuo and the residue was trituated with methanol to give the title compound as crude material which was used in the next steps without further purification (5.9 g, 88% yield).
LC-MS (Method 1 ): Rt = 1.23 min; MS (ESIpos) m/z = 331.2 [M+H]'. Intermediate 18
tert-butyl {4-[(2-chlorophenyl)carbamoyl]phenyl} arbamate
Figure imgf000063_0002
To a mixture of 4- [(tert-butoxycarbonyl)amino] benzoic acid (5.00 g, 20.4 mmol), N,N- diisopropylethylamine (11 ml, 65 mmol) and 1 -[bis(dimethylamino)methylene]-1 H-1 ,2,3- triazolo[4,5-b]-pyridinium 3-oxid hexafluorophosphate (11.7 g, 30.7 mmol) in 65 mL N,N- dimethylformamide was added 2-chloroaniline (3.3 ml, 31 mmol). The mixture was stirred for 3 days at 100 C. For work-up, the reaction mixture was diluted with dichloromethane. The mixture was washed with sodium bicarbonate solution and brine. The combined organic layers were concentrated in vacuo and the residue was trituated with methanol to give the title compound as crude material which was used in the next steps without further purification (2.7 g, 38% yield). LC-MS (Method 1 ): Rt = 1 .33 min; MS (ESI pos) m/z
Intermediate 1
4-amino-N-(2-chloro-4-fluorophenyl)benzamide
Figure imgf000064_0001
To a suspension of tert-butyl {4-[(2-chloro-4-fluorophenyl)carbamoyl] phenyl}carbamate (3.40 g, 9.32 mmol) in dichloromethane (50 ml) was added trifluoroacetic acid (14 ml, 186 mmol) and the mixture was stirred for 5 hours at room temperature. The mixture was concentrated in vacuo and the residue was dissolved in dichloromethane/methanol (9: 1 ). The organic layer was washed with sodium bicarbonate solution and brine. The organic layer was concentrated in vacuo to give the title compound as crude material which was used in the next steps without further purification (2.4 g, 98% yield ).
LC-MS (Method 2): Rt = 1 .00 min; MS (ESI pos) m/z = 265.0 [M+H] '. Intermediate 110
4-amino-N-(4-fluorophenyl)benzamide
Figure imgf000064_0002
To a suspension of tert-butyl {4- [(4-f luorophenyljcarbamoyl] phenyljcarbamate (5.20 g, 1 5.7 mmol) in dichloromethane (81 ml) was added trifluoroacetic acid (24 ml, 310 mmol) and the mixture was stirred for 5 hours at room temperature. The mixture was concentrated in vacuo and the residue was dissolved in dichloromethane/methanol (9: 1 ). The organic layer was washed with sodium bicarbonate solution and brine. The organic layer were concentrated in vacuo to give the title compound as crude material which was used in the next steps without further purification (3.6 g, 99% yield).
LC-MS (Method 2): Rt = 0.89 min; MS (ESIpos) m /z = 231 .1 [M+H] ' . Intermediate 11 1 4-amino-N-(2-chlorophenyl)benzamide
Figure imgf000065_0001
To a suspension of tert-butyl {4-[(2-chlorophenyl)carbamoyl]phenyl}carbamate (2.72 g, 7.84 mmol) in dichloromethane (40 ml) was added trifluoroacetic acid (12 ml, 160 mmol) and the mixture was stirred for 5 hours at room temperature. The mixture was concentrated in vacuo and the residue was dissolved in dichloromethane/methanol (9: 1 ). The organic layer was washed with sodium bicarbonate solution and brine. The organic layer was concentrated in vacuo to give the title compound as crude material which was used in the next steps without further purification (1 .9 g, 99% yield).
LC-MS (Method 2): t = 0.99 min; MS (ESIpos) m/z = 247.1 [M+H] \ Intermediate 112
N,N'-bis{4-[(2-chloro-4-fluorophenyl)carbamoyl]phenyl}-5, 10-dioxo-5H , 10H- diimidazof 1 , 5-a: 1 ', 5'-d]pyrazine- 1 , 6-dicarboxamide
Figure imgf000065_0002
To 5, 10-dioxo-5H , 10H-diimidazo[1 , 5-a: 1 ', 5'-d] pyrazine-1 ,6-dicarbonyl dichloride (716 mg, 2.29 mmol) in tetrahydrofuran (25 ml) were added a solution of 4-amino-N- (2-chloro-4- fluorophenyljbenzamide (1 .21 g, 4.57 mmol) in tetrahydrofuran (25 ml) and N,N- diisopropylethylamine (900 μ , 5.1 mmol). The resulting mixture was stirred for 24 hours at room temperature. The reaction mixture was divided in three portions and each portion was used without further purification in the subsequent steps.
Intermediate 113
N,N'-bis{4-[(4-fluorophenyl)carbamoyl]phenyl}-5, 10-dioxo-5H, 10H-dnmidazo[ 1 , 5- a: 1 ', 5'-d]pyrazine- 1 , 6-dicarboxamide
Figure imgf000066_0001
To 5, 10-dioxo-5H , 10H-diimidazo[1 , 5-a: 1 ', 5 -d] pyrazine- 1 ,6-dicarbonyl dichloride (1.25 g, 3.99 mmol) in tetrahydrofuran (30 ml) were added 4-amino-N-(4-fluorophenyl)benzamide (1.88 g, 7.99 mmol) and N,N-diisopropylethylamine (3.0 ml, 17 mmol). The resulting mixture was stirred for 24 hours at room temperature. The precipitate was collected by filtration and was washed with water (10 ml) and ethylacetate (10 ml). The solids were triturated with methanol to give the title compound (2.2g, 79% yield).
Intermediate 114
N,N'-bis{4-[(2-chlorophenyl)carbamoyl]phenyl}-5, 10-dioxo-5H, 10H-diimidazo[1 ,5- a: 1 ',5'-d]pyrazine- 1 ,6-dicarboxamide
Figure imgf000066_0002
To 5,10-dioxo-5H, 10H-diimidazo[1 ,5-a: 1 ',5'-d]pyrazine-1 ,6-dicarbonyl dichloride (634 mg, 2.03 mmol) in tetrahydrofuran (14 ml) were added 4-amino-N-(2-chloro-phenyl)benzamide (1.00 g, 4.05 mmol) and N,N-diisopropylethylamine (790 μΐ, 4.6 mmol). The resulting mixture was stirred for 24 hours at room temperature. The reaction mixture was divided in three portions and each portion was used without further purification in the subsequent steps.
Intermediate 115
N-(3,5-dichlorophenyl)-4-nitrobenzamide
Figure imgf000067_0001
A mixture of 4-nitrobenzoyl chloride (2.00 g, 10.8 mmol), 3, 5-dichloroaniline (1.75 g, 10.8 mmol) and DMAP (132 mg, 1 .08 mmol) in pyridine (44 ml) was stirred at room temperature over night. For work-up, water was added and the precipitate formed was collected by filtration, washed with water and dried to to give the title compound (2.93 g, 87% yield).
LC-MS (Method 2): t = 1.36 min; MS (ESIpos): m/z = 309.0 [M-H]
1H-NMR (400 MHz, DMS0-d6): δ [ppm] = 10.60 (s, 1 H), 8.42-8.34 (m, 2H), 8.24-8.16 (m, 2H), 7.59 (ddd, 2H), 7.43 (t, 1 H)
Intermediate 116
N-(2,3-dichlorophenyl)-4-nitrobenzamide
Figure imgf000067_0002
Was prepared in analogy to the synthesis of N-(3,5-dichlorophenyl)-4-nitrobenzamide using 4-nitrobenzoyl chloride (2.00 g, 10.8 mmol) and 2,3-dichloroaniline (1 .75 g, 10.8 mmol) as starting materials to give the title compound (2.93g, 87% yield).
LC-MS (Method 2): R. = 1.24 min; MS (ESIneg): m/z = 309.0 [M-H]
H-NMR (400 MHz, DMS0-d5): δ [ppm] = 10.60 (s, 1 H), 8.42-8.34 (m, 2H), 8.24-8.16 (m, 2H), 7.62-756 (m, 2H), 7.43 (t, 1 H)
Intermediate 117
N-(3-chlorophenyl)-4-nitrobenzamide
Figure imgf000068_0001
Was prepared in analogy to the synthesis of N-(3,5-dichlorophenyl)-4-nitrobenzamide using 4-nitrobenzoyl chloride (2.00 g, 10.8 mmol) and 3-chloroaniline (1 .37 g, 10.8 mmol) as starting materials to give the title compound (2.54 g, 85% yield).
LC-MS (Method 2): R- = 1.21 min; MS (ESIneg): m/z = 275.2 [M-H]
'H-NMR (400 MHz, DMS0-d6): δ [ppm] = 10.69 (s, 1 H), 8.41 -8.33 (m, 2H), 8.22-8.12 (m, 2H), 7.95 (t, 1 H), 7.76-7.64 (m, 1 H), 7.40 (t, 1 H ), 7.24-7.15 (m, 1 H)
Intermediate 118
N-[ 1 -(2-chlorophenyl)ethyl]-4-nitrobenzamide
Figure imgf000068_0002
Was prepared in analogy to the synthesis of N-(3,5-dichlorophenyl)-4-nitrobenzamide using 4-nitrobenzoyl chloride (2.00 g, 10.8 mmol) and 1 -(2-chlorophenyl)ethanamine (1 .68 g, 10.8 mmol) as starting materials to give the title compound (2.65 g. 79% yield).
LC-MS (Method 2): R; = 1.19 min; MS (ESIneg): m/z = 303.1 [M-H]
'H-NMR (400 MHz, DMS0-d5): δ [ppm] = 9.26 (d, 1 H ), 8.43-8.24 (m, 2H), 8.20-8.01 (m, 2H), 7.54 (dd, 1 H), 7.42 (dd, 1 H), 7.33 (td, 1 H), 7.26 (td, 1 H ), 5.54-5.33 (m, 1 H), 1 .46 (d, 3H)
Intermediate 119
N-(2,3-dichlorobenzyl)-4-nitrobenzamide
Figure imgf000068_0003
Was prepared in analogy to the synthesis of N-(3,5-dichlorophenyl)-4-nitrobenzamide using 4-nitrobenzoyl chloride (2.00 g. 10.8 mmol) and 1 -(2,3-dichlorophenyl)methanamine (1 .90 g, 10.8 mmol) as starting materials to give the title compound (3.10 g, 86% yield).
LC-MS (Method 2): t = 1.22 min; MS (ESIneg): m/z = 323.0 [M-H]
Ή-NMR (400 MHz, DMS0-d6): δ [ppm] = 9.40 (t, 1 H), 8.36-8.30 (m, 2H), 8.16-8.10 (m, 2H), 7.62-7.53 (m, 1 H), 7.41 -7.29 (m, 2H), 4.59 (d, 2H) intermediate 120
4-amino-N-(3,5-dichlorophenyl)benzamide
Figure imgf000069_0001
Hydrochloric acid (48 ml, 2.0 M aqueous solution, 96 mmol) was added dropwise to a suspension of N-(3,5-dichlorophenyl)-4-nitrobenzamide (3.00 g, 9.64 mmol) and zinc powder (3.15 g, 48.2 mmol) in ethanol (110 ml) under ice-cooling. The mixture was stirred for 2 h at room temperature, then filtrated through a pad of celite, washed with ethanol and the filtrate was concentrated to 1 /3. The residue was poured into water and the mixture was basified with saturated sodium bicarbonate solution. The precipitate formed was collected by filtration, washed with water. The precipitate was dissolved in dichloromethane and the organic phase was washed with water, filtrated through a silicone filter and concentrated to give the title compound (1 .94 g, 72% yield).
LC-MS (Method 2): Rt = 1.19 min; MS (ESIpos): m/z = 281 .0 [M+H] '
Ή-NMR (400 MHz, DMS0-d6): δ [ppm] = 10.03 (s, 1 H), 7.89 (d, 2H), 7.71 (d, 2H), 7.24 (t, 1 H), 6.61 (d, 2H), 5.87 (s, 2H)
Intermediate 121
4-amino-N-(2,3-dichlorophenyl)benzamide
Figure imgf000069_0002
Hydrochloric acid (42 ml, 2.0 M aqueous solution, 84 mmol) was added dropwise to a suspension of N-(2,3-dichlorophenyl)-4-nitrobenzamide (2.60 g, 8.36 mmol) and zinc powder (2.73 g, 41.8 mmol) in ethanol (99 ml) under ice-cooling. The mixture was stirred for 2 h at room temperature, then filtrated through a pad of celite, washed with ethanol and the filtrate was concentrated to 1/3. The residue was extracted with dichloromethane and the organic phase was washed with aqueous sodium hydroxide solution (1 M) and water, filtrated through a silicone filter and concentrated to give the title compound (0.88 g, 37% yield).
LC-MS (Method 2): t = 1.11 min; MS (ESIpos): m/z = 281.0 [M+H]*
Ή-NMR (400 MHz, DMS0-d6): δ [ppm] = 9.59 (s, 1H), 7.79-7.67 (m, 2H), 7.61 (dd, 1H), 7.48 (dd, 1H), 7.36 (t, 1H), 6.65-6.54 (m, 2H), 5.80 (s, 2H)
Intermediate 122
4-amino-N-(3-chlorophenyl)benzamide
Figure imgf000070_0001
Was prepared in analogy to the synthesis of 4-amino-N-(3,5-dichlorophenyl)benzamide using N-(3-chlorophenyl)-4-nitrobenzamide (2.50 g, 9.04 mmol) as starting material to give the title compound (1.00 g, 45% yield).
LC-MS (Method 2): Rt = 1.01 min; MS (ESIpos): m/z = 247.1 [M+H] +
H-NMR (400 MHz, DMS0-d6): δ [ppm] = 9.91 (s, 1H), 7.96 (t, 1H), 7.74-7.66 (m, 3H), 7.34 (t, 1H), 7.13-7.01 (m, 1H), 6.67-6.50 (m, 2H), 5.81 (s, 2H)
Intermediate I23
4-amino-N-[1-(2-chlorophenyl)ethyl]benzamide
Figure imgf000070_0002
Was prepared in analogy to the synthesis of 4-amino-N-(2,3-dichlorophenyl)benzamide using N-[1 -(2-chlorophenyl)ethyl]-4-nitrobenzamide (2.65 g, 8.70 mmol) as starting material to give the title compound (1 .97 g, 82% yield).
LC-MS (Method 2): Rt = 1.22 min; MS (ESIpos): m/z = 275.1 [M+H] +
1H-NMR (400 MHz, DMS0-d6): δ [ppm] = 8.40 (d, 1 H), 7.65-7.59 (m, 2H), 7.50 (dd, 1 H), 7.38 (dd, 1 H), 7.29 (td, 1 H), 7.25-7.17 (m, 1 H), 6.59-6.50 (m, 2H), 5.60 (s, 2H), 5.40 (quin, 1 H), 1 .40 (d, 3H)
Intermediate 124
4-amino-N-(2,3-dichlorobenzyl)benzamide
Figure imgf000071_0001
Was prepared in analogy to the synthesis of 4-amino-N-(2,3-dichlorophenyl)benzamide using N-(2, 3-dichlorobenzyl)-4-nitrobenzamide (3.10 g, 9.53 mmol) as starting material to give the title compound (1 .72 g, 58% yield).
LC-MS (Method 2): Rt = 1.03 min; MS (ESIpos): m/z = 295.1 [M+H] '
'H-NMR (400 MHz, DMS0-d6): δ [ppm] = 8.60 (t, 1 H), 7.62 (d, 2H), 7.53 (dd, 1 H), 7.33 (t, 1 H), 7.29-7.23 (m, 1 H), 6.61 -6.49 (m, 2H), 5.64 (s, 2H), 4.49 (d, 2H)
4. Examples
Example 1
N5-{4-[(3-chlorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin- 1 -yl)ethyl]- 1 H-imidazole- 4, 5-dicarboxamide formic acid salt
x HCOOH
Figure imgf000071_0002
Benzotriazol-1 -yl-oxytripyrrolidinophosphonium hexafluorophosphate (1 17 mg, 225 mol) and N.N-diisopropylethylamine (140 μΐ, 820 mol) were added to a mixture of lithium 4- {[(4-{[2-(piperidin-1-yl)ethy[]carbamoy[}-1H-imidazo[-5-yl)carbony[]amino}benzoate (100 mg, 80 % purity, 204 μιηοί) and 1 -(3-chlorophenyl)methanamine (31.8 mg, 225 μιηοΐ) and the mixture was stirred at room temperature for 12 h. For work-up, the reaction mixture was concentrated and the residue was purified by preparative HPLC (Method 6) followed by recrystallization from ethyl acetate to give the title compound (12.5 mg).
LC-MS (Method 1 ): R, = 0.90 min; MS (ESIpos): m/z = 509.3 [M+H]'
'H-NMR (400 MHz, DMS0-d6) delta [ppm]: 1.156 (0.48), 1.173 (0.97), 1.191 (0.52), 1.372
(0.48), 1.400 (0.61), 1.615 (0.97), 1.647 (1.07), 1.677 (1.03), 1.713 (0.61), 1.729 (0.45),
1.822 (1.10), 1.859 (0.91), 1.907 (0.87), 1.988 (1.84), 2.318 (0.68), 2.323 (1.49), 2.327 (2.04), 2.331 (1.52), 2.337 (0.74), 2.523 (16.00), 2.536 (9.47), 2.540 (8.53), 2.659 (0.71),
2.665 (1.49), 2.669 (2.04), 2.674 (1.45), 2.678 (0.74), 2.923 (0.87), 2.952 (0.94), 3.592
(1.10), 3.624 (1.00), 3.711 (1.26), 3.726 (1.20), 4.474 (3.72), 4.489 (3.75), 7.282 (1.42),
7.286 (1.00), 7.301 (3.20), 7.319 (1.91), 7.324 (2.62), 7.328 (1.45), 7.351 (3.36), 7.364
(2.33), 7.370 (6.14), 7.390 (1.26), 7.759 (3.56), 7.780 (4.36), 7.922 (0.65), 7.942 (4.65), 7.947 (2.20), 7.964 (3.56), 8.035 (2.42), 8.893 (0.45), 9.010 (0.97), 9.026 (1.97), 9.041 (1.00), 9.212 (0.71), 13.612 (1.29), 13.643 (1.36).
Example 2
N5-{4-[(2-chloro-4-fluorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin-1-yl)ethyl]-1H- imidazole-4,5-dicarboxamide formic acid salt
x HCOOH
Figure imgf000072_0001
Was prepared in analogy to the synthesis of N¾-{4-[(3-chlorobenzyl)carbamoyl]phenyl}-N4- [2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4,5-dicarboxamide formic acid salt using lithium 4- {[(4-{[2-(piperidin-1-yl)ethyl]carbamoyl}-1H-imidazol-5-yl)carbonyl]amino}benzoate (100 mg, 80 % purity, 204 μιτιοΐ) and 1-(2-chloro-4-fluorophenyl)methanamine (35.9 mg, 225 pmo) as starting materials to give the title compound (10.5 mg).
LC-MS (Method 1 ): R, = 0.90 min; MS (ESIpos): m/z = 527.3 [M+H]'
'H-NMR (400 MHz, DMS0-d6): δ [ppm] = 13.73-13.4.3 (m, 2H), 9.22 (br. s., 1H), 9.07-8.72 (m, 2H), 8.09-7.88 (m, 3H), 7.81-7.72 (m, 2H), 7.50-7.36 (m, 2H), 7.22 (td, 1H), 4.51 (d, 2H), 3.86-3.50 (m, 4H), 3.02-2.86 (m, 2H), 1.90-1.75 (m, 2H), 1.75-1.51 (m, 3H), 1.48-1.32 (m, 1H)
Example 3
N5-{4-[(3,5-dichlorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin-1-yl)ethyl]-1H- imidazole-4,5-dicarboxamide formic acid salt
x HCOOH
Figure imgf000073_0001
Was prepared in analogy to the synthesis of N5-{4-[(3-chlorobenzyl)carbamoyl]phenyl}-N4- [2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4,5-dicarboxamide formic acid salt using lithium 4- {[(4-{[2-(piperidin-1-yl)ethyl]carbamoyl}-1H-imidazol-5-yl)carbonyl]amino}benzoate (100 mg, 80 % purity, 204 μιτιοΐ) and 1-(3,5-dichlorophenyl)methanamine (39.6 mg, 225 μιηοΐ) as starting materials to give the title compound (23.0 mg).
LC-MS (Method 1): Rt = 0.99 min; MS (ESIpos): m/z = 543.3 [M+H] +
1H-NMR (400 MHz, DMS0-d5): δ [ppm] = 13.78-13.51 (m, 2H), 9.21 (br. s., 1H), 9.05 (t, 1H), 9.00-8.80 (m, 1H), 8.03 (s, 1H), 7.99-7.88 (m, 2H), 7.81-7.72 (m, 2H), 7.50 (t, 1H), 7.37 (d, 2H), 4.48 (d, 2H), 3.84-3.46 (m, 4H), 3.05-2.80 (m, 2H), 1.97-1.29 (m, 6H)
Example 4
N -{4-[(3,5-dichlorophenyl)carbamoyl]phenyl}-N5-[2-(piperidin-1-yl)ethyl]-1H- imidazole-4, 5-dicarboxamide
Figure imgf000073_0002
4-Amino-N-(3,5-dichlorophenyl)benzamide (135 mg. 480 mol) and N,N- diisopropylethylamine (130 μΐ, 720 mol) were added to a suspension of 5,10-dioxo- 5H,10H-diimidazo[1 ,5-a:1',5'-d]pyrazine-1 ,6-dicarbonyl dichloride (75.2 mg, 240 mol) in tetrahydrofuran (4.5 ml) and the mixture was stirred for 6 h at room temperature. Then, a solution of 2-(piperidin-1 -yl)ethanamine (61 .6 mg, 480 μπιοΐ) and N,N- diisopropylethylamine (130 μΐ, 720 pmol) in tetrahydrofuran (3.5 ml) was added and the mixture was stirred for 3 d at room temperature. For work-up, the reaction mixture was concentrated and the residue was purified by preparative HPLC (Method 7) followed by crystallization from methanol to give the title compound (67.0 mg).
LC-MS (Method 2): Rt = 1.37 min; MS (ESIpos): m/z = 529.3 [M+H] +
'H-NMR (400 MHz, DMS0-d6) delta [ppm]: 1 .372 (1 .07), 1 .374 (1 .25), 1 .391 (2.35), 1 .404 (2.20), 1 .418 (1 .10), 1 .484 (1.78), 1 .497 (4.28), 1 .51 1 (5.36), 1 .526 (3.53), 1 .539 (1 .25), 2.317 (0.42), 2.322 (0.90), 2.326 (1.19), 2.331 (0.88), 2.336 (0.48), 2.389 (3.40), 2.404 (4.79), 2.416 (3.38), 2.469 (3.10), 2.522 (3.03), 2.664 (0.88), 2.668 (1.18), 2.673 (0.85), 3.445 (1 .56), 3.461 (3.93), 3.476 (3.82), 3.492 (1.41 ), 7.318 (3.44), 7.323 (7.24), 7.327 (3.62), 7.860 (1 .40), 7.878 (1 .45), 7.894 (1 .32), 7.905 (16.00), 7.910 (14.90), 7.960 (0.40), 7.972 (10.89), 7.992 (5.79), 8.014 (4.1 1 ), 10.457 (5.68), 13.522 (0.57). Example 5
N -{4-[(3, 5-dichlorophenyl)carbamoyl]phenyl}-N5-methyl- 1 H-imidazole-4, 5- dicarboxamide
Figure imgf000074_0001
Was prepared in analogy to the synthesis N4-{4-[(3, 5-dichlorophenyl)carbamoyl]phenyl}-N5- [2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4, 5-dicarboxamide using 5, 10-dioxo-5H, 10H- diimidazo[1 , 5-a: 1 ',5'-d]pyrazine-1 ,6-dicarbonyl dichloride (75.2 mg, 240 pmol), 4-amino-N- (3,5-dichlorophenyl)benzamide (135 mg, 480 pmol) and methanamine (240 μΐ, 2.0 M solution in tetrahydrofuran, 480 pmol) as starting materials. For work-up, the reaction mixture was concentrated and the residue was stirred with DMSO. The precipitate thus formed, was collected by filtration washed with methanol and then recrystallized from methanol to give the title compound (37.7 mg).
LC-MS (Method 1 ): Rt = 1.28 min; MS (ESIpos): m/z = 432.2 [M+H] + 1H-NAAR (400 MHz, DMSO-d6): δ [ppm] = 14.04 (br. s., 1H), 13.51 (br. s., 1H), 10.47 (s, 1 8.92 (br. s., 1H), 8.05-7.99 (m, 2H), 7.98 (s, 1H), 7.92 (d, 2H), 7.91-7.83 (m, 2H), 7.33 1H), 2.89 (d, 3H)
Exampie 6
N -{4-[(2,3-dichlorophenyl)carbamoyl]phenyl}-N5-[2-(piperidin-1-yi)ethyl]-1H- imidazole-4, 5-dicarboxamide
Figure imgf000075_0001
4-Amino-N-(2,3-dichlorophenyl)benzamide (135 mg, 480 μιτιοΐ) and N,N- diisopropylethylamine (130 μΐ, 720 μπιοΐ) were added to a suspension of 5,10-dioxo- 5HJ0H-diimidazo[1,5-a:1',5'-d]pyrazine-1,6-dicarbonyl dichloride (75.2 mg, 240 pmol) in tetrahydrofuran (4.5 ml) and the mixture was stirred for 6 h at room temperature. Then, a solution of 2-(piperidin-1-yl)ethanamine (61.6 mg, 480 μιτιοΐ) and N,N- diisopropylethylamine (130 μΐ, 720 μmol) in tetrahydrofuran (3.5 ml) were added and the mixture was stirred for 3 d at room temperature. For work-up, the reaction mixture was concentrated and the residue was stirred with methanol. The precipitate thus formed was collected by filtration, and purified by preparative HPLC (Method 7) followed by recyrystallization from methanol to give the title compound (5.60 mg).
H-NMR (400 MHz, DMS0-d6): δ [ppm] = 13.95 (br. s., 1H), 13.53 (br. s., 1H), 10.16 (s, 1H), 8.71 (br. s., 1H), 8.08-8.01 (m, 2H), 7.97 (s, 1H), 7.95-7.74 (m, 2H), 7.62-7.54 (m, 2H), 7.47-7.37 (m, 1H), 3.47 (q, 2H), 2.45-2.34 (m, 4H), 1.59-1.45 (m, 4H), 1.44-1.31 (m, 2H)
Example 7
N4-{4-[(3-chlorophenyl)carbamoyl]phenyl}-N5-[2-(piperidin-1-yl)ethyl]-1H-imidazole- 4, 5-dicarboxamide
Figure imgf000076_0001
Was prepared in analogy to the synthesis N4-{4-[(3,5-dichlorophenyl)carbamoyl]phenyl}-N5- [2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4,5-dicarboxamide using 5,10-dioxo-5H,10H- diimidazo[1 ,5-a:r,5'-d]pyrazine-1 ,6-dicarbonyl dichloride (75.2 mg, 240 pmol), 4-amino-N- (3-chlorophenyl)benzamide (118 mg, 480 pmo ) and 2-(piperidin-1 -yl)ethanamine (61.6 mg, 480 pmol) as starting materials to give the title compound (13.0 mg).
LC-MS (Method 5): Rt = 0.88 min; MS (ESIpos): m/z = 495.2 [M+H]+
'H-NMR (400 MHz, DMS0-d6) delta [ppm]: 1.354 (0.42), 1.378 (1 .66), 1.393 (2.97), 1.406 (2.87), 1 .408 (2.76), 1 .423 (1.37), 1.502 (4.66), 1 .516 (5.76), 1 .531 (3.95), 2.317 (0.39), 2.322 (0.74), 2.327 (1.00), 2.332 (0.74), 2.523 (6.00), 2.664 (0.68), 2.669 (0.95), 2.674 (0.71 ), 3.453 (2.08), 3.469 (4.45), 3.484 (4.32), 3.500 (1.82), 7.141 (2.50), 7.142 (2.74), 7.146 (2.84), 7.147 (2.71 ), 7.161 (3.08), 7.163 (3.32), 7.166 (3.42), 7.168 (3.21 ), 7.363 (3.42), 7.370 (0.63), 7.384 (6.29), 7.391 (0.89), 7.404 (3.26), 7.411 (0.45), 7.698 (2.68), 7.700 (3.21 ), 7.705 (3.00), 7.719 (2.58), 7.723 (2.92), 7.726 (2.53), 7.829 (1.95), 7.851 (2.29), 7.928 (0.50), 7.963 (5.45), 7.969 (16.00), 7.983 (2.03), 7.998 (5.00), 8.019 (4.39), 8.114 (0.66), 8.701 (0.92), 10.328 (6.29), 10.357 (0.74), 13.918 (1.37).
Example 8
N -{4-[(3-chiorophenyl)carbamoyl]phenyl}-N5-methyl- 1 H-imidazole-4, 5-dicarboxamide
Figure imgf000076_0002
Was prepared in analogy to the synthesis N4-{4-[(3,5-dichlorophenyl)carbamoyl]phenyl}-N5- [2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4,5-dicarboxamide using 5,10-dioxo-5H,10H- diimidazo[1 ,5-a:1 ',5'-d]pyrazine-1 ,6-dicarbonyl dichloride (75.2 mg, 240 pmol), 4-amino-N- (3-chlorophenyl)benzamide (118 mg, 480 pmol) and methanamine (240 μΐ, 2.0 M solution in tetrahydrofuran, 480 pmol) as starting materials to give the title compound (34.0 mg). LC-MS (Method 5): Rt = 0.1.13 min; MS (ESIpos): m/z = 398.1 [M+H]+
1H-NMR (400 MHz, DMS0-d6): d [ppm] = 13.99 (br. s., 1H), 13.49 (br. s., 1H), 10.32 (s, 1H), 8.88 (br. s., 1H), 8.04-7.91 (m, 4H), 7.90-7.78 (m, 2H), 7.75-7.64 (m, 1H), 7.43-7.32 (m, 1H), 7.21-7.09 (m, 1H), 2.88 (d, 3H) Example 9
N4-{4-[(2,3-dichlorophenyl)carbamoyl]phenyl}-N5-methyl-1H-imidazole-4,5- dicarboxamide
Figure imgf000077_0001
Was prepared in analogy to the synthesis N4-{4-[(3,5-dichlorophenyl)carbamoyl]phenyl}-N5- [2-(piperidin-1-yl)ethyl]-1H-imidazole-4,5-dicarboxamide using 5,10-dioxo-5H,10H- diimidazo[1 ,5-a:1',5'-d]pyrazine-1 ,6-dicarbonyl dichloride (75.2 mg, 240 μιτιοΐ), 4-amino-N- (2,3-dichlorophenyl)benzamide (135 mg, 480 μιτιοΐ) and methanamine (240 μΐ, 2.0 M solution in tetrahydrofuran, 480 mol) as starting materials. For work-up, the reaction mixture was concentrated and the residue was stirred with ethanol. The preicipiated formed was collectec by filtration, washed with water and ethanol and dried to give the title compound (153 mg).
LC-MS (Method 1): R. = 1.19 min; MS (ESIpos): m/z = 432.1 [M+H]'
1H-NMR (400 MHz, DMS0-d6): d [ppm] = 14.01 (s, 1H), 13.50 (br. s., 1H), 10.15 (s, 1H), 8.93-8.85 (m, 1H), 8.04 (d, 2H), 7.96 (s, 1H), 7.84 (d, 2H), 7.59 (dd, 1H), 7.56 (dd, 1H), 7.41 (t, 1H), 2.88 (d, 3H)
Example 10
N4-(4-{[ 1-(2-chlorophenyl)ethyl]carbamoyl}phenyl)-N5-[2-(piperidin- 1 -yl)ethyl]- 1 H- imidazole-4, 5-dicarboxamide
Figure imgf000078_0001
Was prepared in analogy to the synthesis N -{4-[(3, 5-dichlorophenyl)carbamoyl]phenyl}-N5- [2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4, 5-dicarboxamide using 5, 10-dioxo-5H, 10H- diimidazo[1 , 5-a: 1 ',5'-d]pyrazine-1 ,6-dicarbonyl dichloride (150 mg, 479 pmol), 4-amino-N- [-1 -(2-chlorophenyl)ethyl]benzamide (263 mg, 958 μιηοΐ) and 2-(piperidin-1 -yl)ethanamine (123 mg, 958 pmol) as starting materials. For work-up, the reaction mixture was concentrated and the residue was stirred with ethanol. The preicipiated formed was collectec by filtration and purified by preparative HPLC (Method 7) to give the title compound (7.0 mg).
LC-MS (Method 3): R. = 0.95 min; MS (ESIpos): m/z = 523.29 [M+H]'
1H-NMR (400 MHz, DMS0-d6) delta [ppm]: 1 .374 (2.02), 1 .390 (3.73), 1 .403 (3.54), 1 .419 (1 .89), 1 .448 (1 5.88), 1.465 (16.00), 1 .496 (5.98), 1 .51 1 (7.57), 1 .526 (4.76), 1 .907 (0.49), 2.317 (1 .34), 2.322 (2.75), 2.327 (3.73), 2.332 (2.75), 2.336 (1.59), 2.391 (5.80), 2.403 (8.12), 2.417 (5.80), 2.469 (5.92), 2.523 (13.13), 2.539 (4.46), 2.660 (1 .16), 2.664 (2.44), 2.669 (3.54), 2.674 (2.69), 2.679 (1.22), 3.441 (2.44), 3.457 (5.68), 3.472 (5.56), 3.489 (2.20), 5.416 (0.55), 5.433 (2.38), 5.452 (3.66), 5.469 (2.38), 5.487 (0.55), 7.236 (1 .53), 7.240 (1 .65), 7.255 (3.73), 7.259 (3.85), 7.274 (3.1 1 ), 7.278 (3.18), 7.312 (2.32), 7.316 (2.81 ), 7.331 (3.91 ), 7.335 (4.46), 7.350 (1 .95), 7.353 (2.02), 7.406 (5.25), 7.410 (5.13), 7.425 (4.09), 7.429 (4.03), 7.538 (4.09), 7.542 (4.40), 7.557 (3.60), 7.562 (3.54), 7.791 (2.20), 7.932 (6.66), 7.958 (13.62), 8.228 (2.26), 8.677 (0.92), 8.845 (4.09), 8.864 (4.03), 13.838 (1 .16).
Example 1 1
N -(4-{[ 1 -(2-chlorophenyl)ethyl]carbamoyl}phenyl)-N5-methyl- 1 H-imidazole-4, 5- dicarboxamide
Figure imgf000078_0002
Was prepared in analogy to the synthesis N4-{4-[(3,5-dichlorophenyl)carbamoyl]phenyl}-N5- [2-(piperidin-1-yl)ethyl]-1H-imidazole-4,5-dicarboxamide using 5,10-dioxo-5H,10H- diimidazo[1,5-a:1',5'-d]pyrazine-1,6-dicarbonyl dichloride (150 mg, 479 pmol), 4-amino-N- [1-(2-chlorophenyl)ethyl]benzamide (263 mg, 958 pmol) and methanamine (480 μΐ, 2.0 M solution in tetrahydrofuran, 960 pmol) as starting materials. For work-up, the reaction mixture was concentrated and the residue was stirred with ethanol. The preicipiated formed was coUectecd by filtration and purified by preparative HPLC (Method 6) to give the title compound (10.0 mg).
LC-MS (Method 3): Rt = 1.09 min; MS (ESIpos): m/z = 426.2 [M+H]'
Ή-NMR (400 MHz, DMS0-d6) delta [ppm]: 1.426 (13.12), 1.443 (13.15), 2.295 (0.48), 2.300 (1.20), 2.304 (1.65), 2.309 (1.20), 2.314 (0.57), 2.637 (0.54), 2.642 (1.23), 2.646 (1.71), 2.652 (1.20), 2.656 (0.57), 2.852 (14.63), 2.864 (14.77), 5.394 (0.46), 5.412 (1.97), 5.429 (2.97), 5.448 (1.94), 5.465 (0.46), 7.213 (1.37), 7.218 (1.48), 7.232 (3.17), 7.237 (3.17), 7.251 (2.80), 7.255 (2.71), 7.290 (1.85), 7.295 (2.22), 7.310 (3.37), 7.314 (3.48), 7.328 (1.60), 7.332 (1.60), 7.383 (4.51), 7.387 (4.53), 7.403 (3.45), 7.406 (3.48), 7.516 (3.39), 7.521 (3.48), 7.536 (3.05), 7.540 (2.94), 7.754 (0.97), 7.765 (2.11), 7.776 (2.14), 7.788 (2.62), 7.911 (8.41), 7.925 (16.00), 7.933 (6.13), 8.823 (3.54), 8.842 (3.51), 13.449 (0.97).
Example 12
N4-{4-[(2,3-dichlorobenzyl)carbamoyl]phenyl}-N5-[2-(piperidin-1-yl)ethyl]-1H- imidazole-4, 5-dicarboxamide
Figure imgf000079_0001
Was prepared in analogy to the synthesis N4-{4-[(3,5-dichlorophenyl)carbamoyl]phenyl}-N5- [2-(piperidin-1-yl)ethyl]-1H-imidazole-4,5-dicarboxamide using 5,10-dioxo-5H,10H- diimidazo[1 ,5-a:1',5'-d]pyrazine-1 ,6-dicarbonyl dichloride (75.0 mg, 240 pmol), 4-amino-N- (2,3-dichlorobenzyl)benzamide (141 mg, 479 pmol) and 2-(piperidin-1 -yljethanamine (61.4 mg, 479 pmol) as starting materials. For work-up, the reaction mixture was concentrated and the residue was stirred with ethanol. The preicipiated formed was collected by filtration and purified by preparative HPLC (Method 7) to give the title compound (53.0 mg).
LC-MS (Method 1 ): R, = 0.96 min; MS (ESIpos): m/z = 543.3 [M+H] '
'H-NMR (400 MHz, DMS0-d6) delta [ppm]: 1 .372 (1 .71 ), 1 .388 (3.20), 1 .402 (3.01 ), 1 .417 (1 .49), 1 .482 (2.40), 1 .496 (5.70), 1 .509 (7.07), 1 .524 (4.67), 1 .538 (1.71 ), 2.071 (0.64), 2.317 (0.59), 2.322 (1.13), 2.327 (1.54), 2.331 (1.22), 2.336 (0.69), 2.388 (4.99), 2.401 (6.85), 2.415 (4.65), 2.466 (4.04), 2.523 (5.65), 2.539 (1 .20), 2.660 (0.54), 2.664 (0.98), 2.669 (1 .39), 2.674 (1.05), 2.679 (0.46), 3.440 (1.96), 3.457 (5.14), 3.472 (5.04), 3.487 (1 .93), 4.562 (7.27), 4.576 (7.19), 7.322 (0.78), 7.329 (1 .59), 7.342 (7.12), 7.346 (10.45), 7.363 (7.05), 7.383 (2.13), 7.551 (3.91 ), 7.557 (3.43), 7.568 (3.60), 7.575 (3.11 ), 7.827 (1 .98), 7.857 (0.69), 7.944 (6.19), 7.946 (8.10), 7.949 (5.14), 7.956 (16.00), 7.968 (5.97), 9.022 (1 .79), 9.037 (3.79), 9.051 (1.79).
Example 13
N4-{4-[(2,3-dichlorobenzyl)carbamoyl]phenyl}-N5-methyl- 1 H-imidazole-4, 5- dicarboxamide
Figure imgf000080_0001
Was prepared in analogy to the synthesis N4-{4-[(3, 5-dichlorophenyl)carbamoyl]phenyl}-N5- [2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4, 5-dicarboxamide using 5, 10-dioxo-5H, 10H- diimidazo[1 , 5-a: 1 ',5'-d]pyrazine-1 ,6-dicarbonyl dichloride (75.0 mg, 240 μιποΐ), 4-amino-N- (2,3-dichlorobenzyl)benzamide (141 mg, 479 pmol) and methanamine (240 μΐ, 2.0 M solution in tetrahydrofuran, 480 pmol) as starting materials. For work-up, the reaction mixture was concentrated and the residue was stirred with ethanol. The preicipiated formed was collected by filtration and purified by preparative HPLC [Waters Autopurificationsystem: Pump 254, Sample Manager 2767, CFO; colum: XBrigde C18 5 m 100x30 mm; solvent A = water + 0.1 % vol. trifluoroacetic acid; solvent B = acetonitrile; gradient: 0-8 min 36-80% B; flow: 70 ml/min] to give the title compound (18.0 mg).
LC-MS (Method 5): R- = 1.08 min; MS (ESIpos): m/z = 446.1 [M+H] ' 1H-NAAR (400 MHz, DMSO-d6): δ [ppm] = 13.97 (s, 1H), 13.50 (s, 1H), 9.06 (t, 1H), 8.96-8.79 (m, 1H), 8.04-7.86 (m, 3H), 7.81 (d, 2H), 7.57 (dd, 1H), 7.43-7.25 (m, 2H), 4.57 (d, 2H), 2.89 (d, 3H)
Example 14
N5-{4-[(2-chlorophenyl)carbamoyl]phenyl}-N -methyl-1H-imidazole-4,5-dicarboxamide
Figure imgf000081_0001
To a solution of 4-({[4-(methylcarbamoyl)-1H-imidazol-5-yl]carbonyl}amino)benzoic acid (245 mg, 851 pmol) in methanol (24 ml) was added 4-(4,6-dimethoxy-1 ,3,5-triazin-2-yl)-4- methylmorpholin-4-ium chloride (370 mg, 1.28 mmol) and 2-chloroaniline (90 μΐ, 850 μιτιοΐ) and the mixture was first stirred at room temperature for 4 h and then at 50 C for 24 hours. For work-up, the reaction mixture was concentrated and the residue was purified by flash column chromatography followed by trituration first with dichloromethane and then with methanol to give the title compound (35 mg, 11¾yield).
LC-MS (Method 1): Rt = 1.11 min; MS (ESIpos): m/z = 398.0 [M+H] +
1H-NMR (400 MHz, DMS0-d6): δ [ppm] = 14.24 (s, 0.4H), 13.99 (s, 0.4H), 13.61-13.42 (m, 1H), 9.96 (s, 0.5H), 8.96-8.80 (m, 1H), 8.17-7.78 (m, 5H), 7.65-7.51 (m, 1H), 7.41-7.34 (m, 0.5H), 7.32-7.22 (m, 0.5H), 2.88 (d, 3H)
Example 15
N5-{4-[(2-chloro-4-fluorophenyl)carbamoyl]phenyl}-N4-methyl-1H-imidazole-4,5- dicarboxamide
Figure imgf000081_0002
To the crude mixture of N,N'-bis{4-[(2-chloro-4-fluorophenyl)carbamoyl]phenyl}-5,10- dioxo-5H,10H-diimidazo[1 ,5-a:1',5'-d]pyrazine-1 ,6-dicarboxamide (600 mg, 780 mol) in tetrahydrofuran (18 ml) were added methanamine (780 μΐ, 2.0 M in tetrahydrofuran, 1.6 mmol) and triethylamine (270 μΐ, 1.9 mmol) and the mixture was stirred for 3 days at 40 C. The precipitate was collected by filtration and washed with tetrahydrofuran. The solid was then recrystallized from dichloromethane/ methanol (1 :1 ) to give the title compound (290 mg).
LC-MS (Method 1 :): R, = 0.97 min; MS (ESIpos): m/z = 416.1 [M+H]'
'H-NMR (400 MHz, DMS0-d6): δ [ppm] = 14.03 (s, 1 H), 13.52 (s, 1 H), 10.04 (s, 1 H), 8.91 (d, 1 H), 8.12-7.81 (m, 5H), 7.68-7.48 (m, 2H), 7.29 (td, 1 H), 2.89 (d, 3H).
Example 16
N5-{4-[(4-fluorophenyl)carbamoyl]phenyl}-N4-methyl-1 H-imidazole-4, 5-dicarboxamide
Figure imgf000082_0001
To a solution of 4-({[4-(methylcarbamoyl)-1 H-imidazol-5-yl]carbonyl}amino)benzoic acid (200 mg 0.96 mmol) in methanol (20 ml) were added 4-(4,6-dimethoxy-[1 ,3,5]triazin-2-yl)- 4-methylmorpholin-4-ium-chloride (286 mg, 1.04 mmol) and 4-fluoroaniline(77 mg, 0.69 mmol). The mixture was stirred for 16 hours at room temperature. For work-up, the reaction mixture was concentrated in vacuo. Sodium bicarbonate solution was added to the residue (until pH=8 was reached) and the mixture was extracted with ethylacetete (3 times with 50 ml). The combined organic phases were dried over sodium sulfate, and the solvent was removed under reduced pressure. The crude product was purified by flash chromatography to give the title compound (137 mg).
'H-NMR (400 MHz, DMS0-d6): δ [ppm] = 2.89 (d, 3H), 7.17-7.21 (2H), 7.79-7.85 (4H), 7.95- 8.01 (3H), 8.88 (bs, 1 H), 10.22 (s, 1 H), 13.48 (bs, 1 H), 13.97 (bs, 1 H).
Example 17
N4-[2-(dimethylamino)ethyl]-N5-{4-[(4-fluorophenyl)carbamoyl]phenyl}-1 H-imidazole- 4, 5-dicarboxamide
Figure imgf000082_0002
To the crude mixture of N,N'-bis{4-[(4-fluorophenyl)carbamoyl]phenyl}-5, 10-dioxo-5H,10H- diimidazo[1 ,5-a:1 ',5'-d]pyrazine-1 ,6-dicarboxamide (500 mg, 714 μιτιοΐ) in tetrahydrofuran (10 ml) were added N,N-dimethylethane-1 ,2-diamine (160 μΐ, 1.4 mmol) and triethylamine (400 μΐ, 2.9 mmol) and the mixture was stirred for 24 h at room temperature. For work-up, the reaction mixture was concentrated and the residue was purified by flash column chromatography (25 g Snap cartridge, dichloromethane/ methanol-gradient, 20% -> 100% methanol) followed by trituration with dichloromethane to give the title compound (130 mg, 41%yield).
LC-MS (Method 4): Rt = 0.82 min; MS (ES!pos): m/z = 439 [M+H]'
Ή-NMR (400 MHz, DMS0-d6): δ [ppm] = 14.07-13.29 (m, 1 H), 10.24 (s, 1 H), 8.70 (br. s., 1 H), 8.13-7.71 (m, 8H), 7.20 (t, 2H), 3.46 (d, 2H), 2.46 (br. s. , 2H), 2.21 (s, 6H).
Example 18
N5-{4-[(4-fluorophenyl)carbamoyl]phenyl}-N -[2-(piperidin-1-yl)ethyl]-1 H-imidazole- 4, 5-dicarboxamide
Figure imgf000083_0001
To the crude mixture of N,N'-bis{4-[(4-fluorophenyl)carbamoyl]phenyl}-5, 10-dioxo-5H,10H- diimidazo[1 ,5-a:1 ',5'-d]pyrazine-1 ,6-dicarboxamide (500 mg, 714 pmol) in tetrahydrofuran (10 ml) were added 2-(piperidin-1 -yl)ethanamine (200 μΐ, 1.4 mmol) and triethylamine (400 μΐ, 2.9 mmol) and the mixture was stirred for 24 h at room temperature. For work-up, the reaction mixture was concentrated and the residue was purified by flash column chromatography (25 g Snap cartridge, dichloromethane/ methanol-gradient, 20% -> 100% methanol) followed by trituration with dichloromethane to give the title compound (56 mg, 17 % yield).
LC-MS (Method 2): Rt = 1.1 min; MS (ESIpos): m/z = 479.2 [M+H]'
'H-NMR (400 MHz, DMS0-d6): δ [ppm] = 13.92 (br. s. , 1 H), 13.54 (br. s. , 1 H), 10.24 (s, 1 H), 8.71 (br. s. , 1 H), 8.11 -7.65 (m, 7H), 7.31 -7.10 (m, 2H), 3.48 (q, 2H), 2.45-2.29 (m, 4H), 1.68-1.17 (m, 6H).
Example 19 N5-{4-[(2-chloro-4-fluorophenyl)carbamoyl]phenyl}-N -[2-(piperidin- 1 -yl)ethyl]-1 H- imidazole-4, 5-dicarboxamide
Figure imgf000084_0001
To the crude mixture of N,N'-bis{4-[(2-chloro-4-fluorophenyl)carbamoyl]phenyl}-5, 10- dioxo-5H, 10H-diimidazo[1 ,5-a: 1 ',5'-d]pyrazine-1 ,6-dicarboxamide (600 mg, 780 μιτιοΐ) in tetrahydrofuran (10 ml) were added 2-(piperidin-1 -yljethanamine (220 μΐ, 1 .6 mmol) and triethylamine (270 μΐ, 1 .9 mmol) and the mixture was stirred for 24 hours at room temperature. The precipitate was collected by filtration and washed with tetrahydrofuran and was then recrystallized from dichloromethane/methanol to give the title compound (376 mg, 89 % yield).
LC-MS (Method 4): Rt = 0.90 min; MS (ESIpos): m/z = 513 [M+H] +
'H-NMR (400 MHz, DMS0-d6): δ [ppm] = 13.92 (br. s. , 1 H), 13.55 (br. s. , 1 H), 10.14-9.99 (m, 1 H), 8.75 (br. s. , 1 H), 8.14-7.79 (m, 5H), 7.67-7.49 (m, 2H ), 7.37-7.17 (m, 1 H), 3.50 (d, 2H), 2.47 (br. s. , 2H), 1 .63-1.27 (m, 6H). Example 20
N5-{4-[(2-chloro-4-fluorophenyl)carbamoyl]phenyl}-N4-[2-(dimethylamino)ethyl]- 1 H- imidazole-4, 5-dicarboxamide
Figure imgf000084_0002
To the crude mixture of N,N'-bis{4-[(2-chloro-4-fluorophenyl)carbamoyl]phenyl}-5, 10- dioxo-5H, 10H-diimidazo[1 ,5-a: 1 ',5'-d]pyrazine-1 ,6-dicarboxamide (600 mg, 780 pmol) in tetrahydrofuran (10 ml) was added N, N-dimethylethane-1 ,2-diamine (170 μΐ, 1 .6 mmol) and triethylamine (270 μΐ, 1 .9 mmol) and the mixture was stirred for 24 h at room temperature. The precipitate was collected by filtration and washed with tetrahydrofuran and was then recrystallized from dichloromethane/methanol to give the title compound (339 mg, 89 % yield).
LC-MS (Method 2): Rt = 1.04 min; MS (ES!pos): m/z = 473.1 [M+H] ' 1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 13.91 (br. s., 1H), 10.03 (s, 1H), 8.71 (br. s., 1H), 8.10-7.78 (m, 5H), 7.66-7.54 (m, 2H), 7.29 (td, 1H), 3.48 (q, 2H), 2.24 (s, 6H).
Example 21
N5-{4-[(2-chlorophenyl)carbamoyl]phenyl}-N4-[2-(piperidin-1-yl)ethyl]-1H-imidazole- 4, 5-dicarboxamide
Figure imgf000085_0001
To the crude mixture of N,N'-bis{4-[(2-chlorophenyl)carbamoyl]phenyl}-5,10-dioxo-5H,10H- diimidazo[1,5-a:1\5'-d]pyrazine-1 ,6-dicarboxamide (750 mg, 1.02 mmol) in tetrahydrofuran (25 ml) was added 2-(piperidin-1 -yl)ethanamine (290 μΐ, 2.0 mmol) and triethylamine (360 μΐ, 2.6 mmol) and the mixture was stirred for 28 h at room temperature. For work-up, the reaction mixture was concentrated and the residue was purified by flash column chromatography (25 g Snap cartridge, dichloromethane/ methanol-gradient, 20% -> 100% methanol) followed by trituration with dichloromethane to give the title compound (417 mg, 80 % yield).
LC-MS (Method 1): Rt = 0.91 min; MS (ESIpos): m/z = 495.3 [M+H]'
1H-NMR (400 MHz, DMS0-d6): δ [ppm] = 13.87 (br. s., 1H), 13.59 (br. s., 1H), 10.00 (s, 1H), 8.14-7.78 (m, 5H), 7.61 (dd, 1H), 7.57 (dd, 1H) 7.40 (td, 1H), 7.36-7.22 (m, 1H), 3.56 (br. s., 2H), 1.88-1.11 (m, 6H).
Example 22
N5-{4-[(2-chlorophenyl)carbamoyl]phenyl}-N -[2-(dimethylamino)ethyl]-1H-imidazole- 4, 5-dicarboxamide
Figure imgf000085_0002
To the crude mixture of N,N'-bis{4-[(2-chlorophenyl)carbamoyl]phenyl}-5,10-dioxo-5H,10H- diimidazo[1,5-a:1',5'-d]pyrazine-1 ,6-dicarboxamide (750 mg, 1.02 mmol) in tetrahydrofuran (25 ml) was added N,N-dimethylethane-1 ,2-diamine (220 μΐ, 2.0 mmol) and triethylamine (360 μΐ, 2.6 mmol) and the mixture was stirred for 28 h at room temperature. The precipitate was collected by filtration and washed with tetrahydrofuran and was then recrystallized from dichloromethane/ methanol to give the title compound (345 mg, 74 % yield).
LC-MS (Method 2): Rt = 1.01 min; MS (ESIpos): m/z = 455.2 [M+H] '
1H-NMR (400 MHz, DMS0-d6): δ [ppm] = 13.90 (br. s. , 1 H), 13.56 (br. s. , 1 H), 9.99 (s, 1 H), 8.75 (br. s. , 1 H), 8.12-7.78 (m, 5H), 7.69-7.49 (m, 2H), 7.44-7.36 (m, 1 H), 7.30 (td, 1 H), 3.50 (q, 2H), 2.58 (br. s. , 2H), 2.38-2.21 (m, 6H ).
Further, the compounds of formula (I) of the present invention can be converted to any salt as described herein, by any method which is known to the person skilled in the art. Similarly, any salt of a compound of formula (I) of the present invention can be converted into the free compound, by any method which is known to the person skilled in the art. Pharmaceutical compositions of the compounds of the invention
This invention also relates to pharmaceutical compositions containing one or more compounds of the present invention. These compositions can be utilised to achieve the desired pharmacological effect by administration to a patient in need thereof. A patient, for the purpose of this invention, is a mammal, including a human, in need of treatment for the particular condition or disease. Therefore, the present invention includes pharmaceutical compositions that are comprised of a pharmaceutically acceptable carrier and a pharmaceutically effective amount of a compound, or salt thereof, of the present invention. A pharmaceutically acceptable carrier is preferably a carrier that is relatively non-toxic and innocuous to a patient at concentrations consistent with effective activity of the active ingredient so that any side effects ascribable to the carrier do not vitiate the beneficial effects of the active ingredient. A pharmaceutically effective amount of compound is preferably that amount which produces a result or exerts an influence on the particular condition being treated. The compounds of the present invention can be administered with pharmaceutically-acceptable carriers well known in the art using any effective conventional dosage unit forms, including immediate, slow and timed release preparations, orally, parenterally, topically, nasally, ophthalmically, optically, sublingually, rectally, vaginally, and the like.
For oral administration, the compounds can be formulated into solid or liquid preparations such as capsules, pills, tablets, troches, lozenges, melts, powders, solutions, suspensions, or emulsions, and may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions. The solid unit dosage forms can be a capsule that can be of the ordinary hard- or soft-shelled gelatine type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and corn starch.
In another embodiment, the compounds of this invention may be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatine, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, gum tragacanth, acacia, lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid, or magnesium, calcium or zinc stearate, dyes, colouring agents, and flavouring agents such as peppermint, oil of wintergreen, or cherry flavouring, intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient. Suitable excipients for use in oral liquid dosage forms include dicalcium phosphate and diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both.
Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example those sweetening, flavouring and colouring agents described above, may also be present.
The pharmaceutical compositions of this invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil such as liquid paraffin or a mixture of vegetable oils. Suitable emulsifying agents may be (1 ) naturally occurring gums such as gum acacia and gum tragacanth, (2) naturally occurring phosphatides such as soy bean and lecithin, (3) esters or partial esters derived form fatty acids and hexitol anhydrides, for example, sorbitan monooleate, (4) condensation products of said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavouring agents. Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil such as, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent such as, for example, beeswax, hard paraffin, or cetyl alcohol. T e suspensions may also contain one or more preservatives, for example, ethyl or n-propyl p- hydro xybenzoate ; one or more colouring agents ; one or more flavouring agents ; and one or more sweetening agents such as sucrose or saccharin.
Syrups and elixirs may be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, and preservative, such as methyl and propyl parabens and flavouring and colouring agents.
The compounds of this invention may also be administered parenterally, that is, subcutaneously, intravenously, intraocularly, intrasynovially, intramuscularly, or interperitoneally, as injectable dosages of the compound in preferably a physiologically acceptable diluent with a pharmaceutical carrier which can be a sterile liquid or mixture of liquids such as water, saline, aqueous dextrose and related sugar solutions, an alcohol such as ethanol, isopropanol, or hexadecyl alcohol, glycols such as propylene glycol or polyethylene glycol, glycerol ketals such as 2,2-dimethyl-1 ,1 -dioxolane-4-methanol, ethers such as polyethylene glycol) 400, an oil, a fatty acid, a fatty acid ester or, a fatty acid glyceride, or an acetylated fatty acid glyceride, with or without the addition of a pharmaceutically acceptable surfactant such as a soap or a detergent, suspending agent such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agent and other pharmaceutical adjuvants.
Illustrative of oils which can be used in the parenteral formulations of this invention are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, sesame oil, cottonseed oil, corn oil, olive oil, petrolatum and mineral oil. Suitable fatty acids include oleic acid, stearic acid, isostearic acid and myristic acid. Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate. Suitable soaps include fatty acid alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamine acetates ; anionic detergents, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates ; non-ionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and poly(oxyethylene-oxypropylene)s or ethylene oxide or propylene oxide copolymers ; and amphoteric detergents, for example, alkyl-beta-aminopropionates, and 2-alkylimidazoline quaternary ammonium salts, as well as mixtures.
The parenteral compositions of this invention will typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimise or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile- lipophile balance (HLB) preferably of from about 12 to about 17. The quantity of surfactant in such formulation preferably ranges from about 5% to about 15% by weight. The surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB.
Illustrative of surfactants used in parenteral formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
The pharmaceutical compositions may be in the form of sterile injectable aqueous suspensions. Such suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents such as, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia ; dispersing or wetting agents which may be a naturally occurring phosphatide such as lecithin, a condensation product of an alkylene oxide with a fatty acid, for example, polyoxyethylene stearate, a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadeca- ethyleneoxycetanol, a condensation product of ethylene oxide with a partial ester derived form a fatty acid and a hexitol such as polyoxyethylene sorbitol monooleate, or a condensation product of an ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride, for example polyoxyethylene sorbitan monooleate.
The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Diluents and solvents that may be employed are, for example, water, Ringer's solution, isotonic sodium chloride solutions and isotonic glucose solutions. In addition, sterile fixed oils are conventionally employed as solvents or suspending media. For this purpose, any bland, fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid can be used in the preparation of injectables.
A composition of the invention may also be administered in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are, for example, cocoa butter and polyethylene glycol.
Another formulation employed in the methods of the present invention employs transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art (see, e.g. , US Patent No. 5,023,252, issued June 1 1 . 1991 , incorporated herein by reference). Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
Controlled release formulations for parenteral administration include liposomal, polymeric microsphere and polymeric gel formulations that are known in the art.
It may be desirable or necessary to introduce the pharmaceutical composition to the patient via a mechanical delivery device. The construction and use of mechanical delivery devices for the delivery of pharmaceutical agents is well known in the art. Direct techniques for, for example, administering a drug directly to the brain usually involve placement of a drug delivery catheter into the patient's ventricular system to bypass the blood-brain barrier. One such implantable delivery system, used for the transport of agents to specific anatomical regions of the body, is described in US Patent No. 5,01 1 ,472, issued April 30, 1991 .
The compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired. Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized. Such ingredients and procedures include those described in the following references, each of which is incorporated herein by reference: Powell, M. F. et al. , "Compendium of Excipients for Parenteral Formulations" PDA Journal of Pharmaceutical Science 6t Technology 1998, 52(5), 238-31 1 ; Strickley, R.G "Parenteral Formulations of Small Molecule Therapeutics Marketed in the United States (1999)-Part-1 " PDA Journal of Pharmaceutical Science & Technology 1999, 53(6), 324-349 ; and Nema, S. et al. , "Excipients and Their Use in Injectable Products" PDA Journal of Pharmaceutical Science & Technology 1997, 51 (4), 166-171 .
Commonly used pharmaceutical ingredients that can be used as appropriate to formulate the composition for its intended route of administration include: acidifying agents (examples include but are not limited to acetic acid, citric acid, fumaric acid, hydrochloric acid, nitric acid) ;
alkali nizing agents (examples include but are not limited to ammonia solution, ammonium carbonate, diethanolamine, monoethanolamine, potassium hydroxide, sodium borate, sodium carbonate, sodium hydroxide, triethanolamine, trolamine) ;
adsorbents (examples include but are not limited to powdered cellulose and activated charcoal) ;
aerosol propellents (examples include but are not limited to carbon dioxide, CCI2F2, F2C C- CClFz and CCIF3)
air displacement agents (examples include but are not limited to nitrogen and argon) ; antifungal preservatives (examples include but are not limited to benzoic acid, butylparaben, ethylparaben, methylparaben, propylparaben, sodium benzoate) ;
antimicrobial preservatives (examples include but are not limited to benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetylpyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, phenylmercuric nitrate and thimerosal) ;
antioxidants (examples include but are not limited to ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorus acid, monothioglycerol, propyl gallate, sodium ascorbate, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite) ;
binding materials (examples include but are not limited to block polymers, natural and synthetic rubber, polyacrylates, polyurethanes, silicones, polysiloxanes and styrene- butadiene copolymers) ;
buffering agents (examples include but are not limited to potassium metaphosphate, dipotassium phosphate, sodium acetate, sodium citrate anhydrous and sodium citrate dihydrate)
carrying agents (examples include but are not limited to acacia syrup, aromatic syrup, aromatic elixir, cherry syrup, cocoa syrup, orange syrup, syrup, corn oil, mineral oil, peanut oil, sesame oil, bacteriostatic sodium chloride injection and bacteriostatic water for injection)
chelating agents (examples include but are not limited to edetate disodium and edetic acid) colourants (examples include but are not limited to FD&C Red No. 3, FD&C Red No. 20, FD&C Yellow No. 6, FD&C Blue No. 2, D&C Green No. 5, D&C Orange No. 5, D&C Red No. 8, caramel and ferric oxide red) ;
clarifying agents (examples include but are not limited to bentonite) ;
emulsifying agents (examples include but are not limited to acacia, cetomacrogol, cetyl alcohol, glyceryl monostearate, lecithin, sorbitan monooleate, polyoxyethylene 50 monostearate) ;
encapsulating agents (examples include but are not limited to gelatin and cellulose acetate phthalate)
flavourants (examples include but are not limited to anise oil, cinnamon oil, cocoa, menthol, orange oil, peppermint oil and vanillin) ;
humectants (examples include but are not limited to glycerol, propylene glycol and sorbitol) ;
levigating agents (examples include but are not limited to mineral oil and glycerin) ;
oils (examples include but are not limited to arachis oil, mineral oil, olive oil, peanut oil, sesame oil and vegetable oil) ;
ointment bases (examples include but are not limited to lanolin, hydrophilic ointment, polyethylene glycol ointment, petrolatum, hydrophilic petrolatum, white ointment, yellow ointment, and rose water ointment) ;
penetration enhancers (transdermal delivery) (examples include but are not limited to monohydroxy or polyhydroxy alcohols, mono-or polyvalent alcohols, saturated or unsaturated fatty alcohols, saturated or unsaturated fatty esters, saturated or unsaturated dicarboxylic acids, essential oils, phosphatidyl derivatives, cephalin, terpenes, amides, ethers, ketones and ureas)
plasticizers (examples include but are not limited to diethyl phthalate and glycerol) ; solvents (examples include but are not limited to ethanol, corn oil, cottonseed oil, glycerol, isopropanol, mineral oil, oleic acid, peanut oil, purified water, water for injection, sterile water for injection and sterile water for irrigation) ;
stiffening agents (examples include but are not limited to cetyl alcohol, cetyl esters wax, microcrystalline wax, paraffin, stearyl alcohol, white wax and yellow wax) ;
suppository bases (examples include but are not limited to cocoa butter and polyethylene glycols (mixtures)) ; surfactants (examples include but are not limited to benzalkonium chloride, nonoxynol 10, oxtoxynol 9, polysorbate 80, sodium lauryl sulfate and sorbitan mono-palmitate) ;
suspending agents (examples include but are not limited to agar, bentonite, carbomers, carboxymethylcellulose sodium, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, kaolin, methylcellulose, tragacanth and veegum) ;
sweetening agents (examples include but are not limited to aspartame, dextrose, glycerol, mannitol, propylene glycol, saccharin sodium, sorbitol and sucrose) ;
tablet anti-adherents (examples include but are not limited to magnesium s tea rate and talc) ;
tablet binders (examples include but are not limited to acacia, alginic acid, carboxymethylcellulose sodium, compressible sugar, ethylcellulose, gelatin, liquid glucose, methylcellulose, non-crosslinked polyvinyl pyrrolidone, and pregelatinized starch) ;
tablet and capsule diluents (examples include but are not limited to dibasic calcium phosphate, kaolin, lactose, mannitol, microcrystalline cellulose, powdered cellulose, precipitated calcium carbonate, sodium carbonate, sodium phosphate, sorbitol and starch) ;
tablet coating agents (examples include but are not limited to liquid glucose, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose, ethylcellulose, cellulose acetate phthalate and shellac) ;
tablet direct compression excipients (examples include but are not limited to dibasic calcium phosphate) ;
tablet disintegrants (examples include but are not limited to alginic acid, carboxymethylcellulose calcium, microcrystalline cellulose, polacrillin potassium, cross- linked polyvinylpyrrolidone, sodium alginate, sodium starch glycollate and starch) ;
tablet glidants (examples include but are not limited to colloidal silica, corn starch and talc) ;
tablet lubricants (examples include but are not limited to calcium stearate, magnesium stearate, mineral oil, stearic acid and zinc stearate) ;
tablet/capsule opaquants (examples include but are not limited to titanium dioxide) ; tablet polishing agents (examples include but are not limited to carnuba wax and white wax) ; thickening agents (examples include but are not limited to beeswax , cetyl alcohol and paraffin) ;
tonicity agents (examples include but are not limited to dextrose and sodium chloride) ; viscosity increasing agents (examples include but are not limited to alginic acid, bentonite, carbomers, carboxymethylcellulose sodium, methylcellulose, polyvinyl pyrrolidone, sodium alginate and tragacanth) ; and
wetting agents (examples include but are not limited to heptadecaethylene oxycetanol, lecithins, sorbitol monooleate, polyoxyethylene sorbitol monooleate, and polyoxyethylene stearate).
Pharmaceutical compositions according to the present invention can be illustrated as follows:
Sterile IV Solution: A 5 mg/mL solution of the desired compound of this invention can be made using sterile, injectable water, and the pH is adjusted if necessary. The solution is diluted for administration to 1 - 2 mg/mL with sterile 5% dextrose and is administered as an IV infusion over about 60 min.
Lvophi ised powder for IV administration: A sterile preparation can be prepared with (i) 100 - 1000 mg of the desired compound of this invention as a lyophilised powder, (ii) 32- 327 mg/mL sodium citrate, and (iii) 300 - 3000 mg Dextran 40. The formulation is reconstituted with sterile, injectable saline or dextrose 5% to a concentration of 10 to 20 mg/mL, which is further diluted with saline or dextrose 5% to 0.2 - 0.4 mg/mL, and is administered either IV bolus or by IV infusion over 15 60 min.
Intramuscular suspension: The following solution or suspension can be prepared, for intramuscular injection:
50 mg/mL of the desired, water-insoluble compound of this invention
5 mg/mL sodium carboxymethylcellulose
4 mg/mL TWEEN 80
9 mg/mL sodium chloride
9 mg/mL benzyl alcohol
Hard Shell Capsules: A large number of unit capsules are prepared by filling standard two- piece hard galantine capsules each with 100 mg of powdered active ingredient, 150 mg of lactose, 50 mg of cellulose and 6 mg of magnesium stearate. Soft Gelatin Capsules: A mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil is prepared and injected by means of a positive displacement pump into molten gelatin to form soft gelatin capsules containing 100 mg of the active ingredient. The capsules are washed and dried. The active ingredient can be dissolved in a mixture of polyethylene glycol, glycerin and sorbitol to prepare a water miscible medicine mix.
Tablets: A large number of tablets are prepared by conventional procedures so that the dosage unit is 100 mg of active ingredient, 0.2 mg. of colloidal silicon dioxide, 5 mg of magnesium stearate, 275 mg of microcrystalline cellulose, 1 1 mg of starch, and 98.8 mg of lactose. Appropriate aqueous and non-aqueous coatings may be applied to increase palatability, improve elegance and stability or delay absorption.
Immediate Release Tablets/ Capsules: These are solid oral dosage forms made by conventional and novel processes. These units are taken orally without water for immediate dissolution and delivery of the medication. The active ingredient is mixed in a liquid containing ingredient such as sugar, gelatin , pectin and sweeteners. These liquids are solidified into solid tablets or caplets by freeze drying and solid state extraction techniques. The drug compounds may be compressed with viscoelastic and thermoelastic sugars and polymers or effervescent components to produce porous matrices intended for immediate release, without the need of water.
Combination therapies
The term "combination" in the present invention is used as known to persons skilled in the art and may be present as a fixed combination, a non-fixed combination or kit-of-parts. A "fixed combination" in the present invention is used as known to persons skilled in the art and is defined as a combination wherein the said first active ingredient and the said second active ingredient are present together in one unit dosage or in a single entity. One example of a "fixed combination" is a pharmaceutical composition wherein the said first active ingredient and the said second active ingredient are present in admixture for simultaneous administration, such as in a formulation. Another example of a "fixed combination" is a pharmaceutical combination wherein the said first active ingredient and the said second active ingredient are present in one unit without being in admixture. A non-fixed combination or "kit-of-parts" in the present invention is used as known to persons skilled in the art and is defined as a combination wherein the said first active ingredient and the said second active ingredient are present in more than one unit. One example of a non-fixed combination or kit-of-parts is a combination wherein the said first active ingredient and the said second active ingredient are present separately. The components of the non-fixed combination or kit-of-parts may be administered separately, sequentially, simultaneously, concurrently or chronologically staggered.
The compounds of this invention can be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutical agents where the combination causes no unacceptable adverse effects. The present invention relates also to such combinations. For example, the compounds of this invention can be combined with known chemotherapeutic agents or anti-cancer agents, e.g. anti-hyper-proliferative or other indication agents, and the like, as well as with admixtures and combinations thereof. Other indication agents include, but are not limited to, anti-angiogenic agents, mitotic inhibitors, alkylating agents, anti-metabolites, DNA-intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzyme inhibitors, toposisomerase inhibitors, biological response modifiers, or anti-hormones. The term "chemotherapeutic anti-cancer agents", includes but is not limited to
131 1-chTNT, abarelix, abiraterone, aclarubicin, ado-trastuzumab emtansine, afatinib, aflibercept, aldesleukin, alectinib, alemtuzumab, alendronic acid, alitretinoin, altretamine, amifostine, aminoglutethimide, hexyl aminolevulinate, amrubicin, amsacrine, anastrozole, ancestim, anethole dithiolethione, anetumab ravtansine, angiotensin II, antithrombin III, aprepitant, arcitumomab, arglabin, arsenic trioxide, asparaginase, axitinib, azacitidine, basiliximab, belotecan, bendamustine, besilesomab, belinostat, bevacizumab, bexarotene, bicalutamide, bisantrene, bleomycin, blinatumomab, bortezomib, buserelin, bosutinib, brentuximab vedotin, busulfan, cabazitaxel, cabozantinib, calcitonine, calcium folinate, calcium levofolinate, capecitabine, capromab, carboplatin, carboquone, carfilzomib, carmofur, carmustine, catumaxomab, celecoxib, celmoleukin, ceritinib, cetuximab, chlorambucil, chlormadinone, chlormethine, cidofovir, cinacalcet, cisplatin, cladribine, clodronic acid, clofarabine, cobimetinib, copanlisib , crisantaspase, crizotinib, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, daratumumab, darbepoetin a If a, dabrafenib, dasatinib, daunorubicin, decitabine, degarelix, den i leu kin diftitox, denosumab, depreotide, deslorelin, dianhydrogalactitol, dexrazoxane, dibrospidium chloride, dianhydrogalactitol, diclofenac, dinutuximab, docetaxel, dolasetron , doxifluridine, doxorubicin, doxorubicin + estrone, dronabinol, eculizumab, edrecolomab, elliptinium acetate, elotuzumab, eltrombopag, endostatin, enocitabine, enzalutamide, epirubicin , epitiostanol, epoetin alfa, epoetin beta, epoetin zeta, eptaplatin , eribulin , erlotinib , esomeprazole, estradiol, estramustine, ethinylestradiol, etoposide, everolimus, exemestane, fadrozole, fentanyl, filgrastim, fluoxymesterone, floxuridine, fludarabine, fluorouracil , flutamide, folinic acid, formestane, fosaprepitant, fotemustine, fulvestrant, gadobutrol, gadoteridol, gadoteric acid meglumine, gadoversetamide, gadoxetic acid, gallium nitrate, ganirelix , gefitinib, gemcitabine, gemtuzumab, Glucarpidase, glutoxim , GM-CSF, goserelin, granisetron, granulocyte colony stimulating factor, histamine dihydrochloride, histrelin , hydroxycarbamide, 1-125 seeds, lansoprazole, ibandronic acid, ibritumomab tiuxetan, ibrutinib, idarubicin, ifosfamide, imatinib, imiquimod , improsulfan , indisetron , incadronic acid, ingenol mebutate, interferon alfa, interferon beta, interferon gamma, iobitridol, iobenguane (1231 ), iomeprol, ipilimumab, irinotecan , Itraconazole, ixabepilone, ixazomib, lanreotide, lansoprazole, lapatinib, lasocholine, lenalidomide, lenvatinib, lenograstim , lentinan , letrozole, leuprorelin , leva mi sole, levonorgestrel, levothyroxine sodium , lisuride, lobaplatin , lomustine, lonidamine, masoprocol, medroxyprogesterone, megestrol, melarsoprol, melphalan , mepitiostane, mercaptopurine, mesna , methadone, methotrexate, methoxsalen , methylaminolevulinate, methylprednisolone, methyl testosterone, metirosine, mifamurtide, miltefosine, miriplatin, mitobronitol, mitoguazone, mitolactol, mitomycin , mitotane, mitoxantrone, mogamulizumab, molgramostim , mopidamol, morphine hydrochloride, morphine sulfate, nabilone, nabiximols, nafarelin , naloxone + pentazocine, naltrexone, nartograstim, necitumumab, nedaplatin , nelarabine, neridronic acid, netupitant/palonosetron , nivolumabpentetreotide, nilotinib, nilutamide, nimorazole, nimotuzumab, nimustine, nintedanib, nitracrine, nivolumab, obinutuzumab, octreotide, ofatumumab, olaparib, omacetaxine mepesuccinate, omeprazole, ondansetron , oprelvekin, orgotein , orilotimod, osimertinib, oxaliplatin, oxycodone, oxymetholone, ozogamicine, p53 gene therapy, paclitaxel, palbociclib, palifermin , palladium-103 seed, palonosetron , pamidronic acid, panitumumab, panobinostat, pantoprazole, pazopanib, pegaspargase, PEG-epoetin beta (methoxy PEG-epoetin beta) , pembrolizumab, pegfilgrastim, peginterferon alfa-2b, pemetrexed, pentazocine, pentostatin , peplomycin , Perflubutane, perfosfamide, Pertuzumab, picibanil, pilocarpine, pirarubicin, pixantrone, plerixafor, plicamycin, poliglusam, polyestradiol phosphate, polyvinylpyrrolidone + sodium hyaluronate, polysaccharide-K, pomalidomide, ponatinib, porfimer sodium, pralatrexate, prednimustine, prednisone, procarbazine, procodazole, propranolol, quinagolide, rabeprazole, racotumomab, radium-223 chloride, radotinib, raloxifene, raltitrexed, ramosetron, ramucirumab, ranimustine, rasburicase, razoxane, refametinib , regorafenib, risedronic acid, rhenium-186 etidronate, rituximab, rolapitant, romidepsin, romiplostim, romurtide, roniciclib , samarium (153Sm) lexidronam, sargramostim, satumomab, secretin, siltuximab, sipuleucel-T, sizofiran, sobuzoxane, sodium glycididazole, sonidegib, sorafenib, stanozolol, streptozocin, sunitinib, talaporfin, talimogene laherparepvec, tamibarotene, tamoxifen, tapentadol, tasonermin, teceleukin, technetium (99mTc) nofetumomab merpentan, 99mTc-HYNIC-[Tyr3]-octreotide, tegafur, tegafur + gimeracil + oteracil, temoporfin, temozolomide, temsirolimus, teniposide, testosterone, tetrofosmin, thalidomide, thiotepa, thymalfasin, thyrotropin a If a, tioguanine, tocilizumab, topotecan, toremifene, tositumomab, trabectedin, trametinib, tramadol, trastuzumab, trastuzumab emtansine, treosulfan, tretinoin, trifluridine + tipiracil, trilostane, triptorelin, trametinib, trofosfamide, thrombopoietin, tryptophan, ubenimex, valatinib , valrubicin, vandetanib, vapreotide, vemurafenib, vinblastine, vincristine, vindesine, vinflunine, vinorelbine, vismodegib, vorinostat, vorozole, yttrium-90 glass microspheres, zinostatin, zinostatin stimalamer, zoledronic acid, zorubicin.
The compounds of the invention may also be administered in combination with protein therapeutics. Such protein therapeutics suitable for the treatment of cancer or other angiogenic disorders and for use with the compositions of the invention include, but are not limited to, an interferon (e.g. , interferon .alpha. , .beta. , or .gamma. ) supraagonistic monoclonal antibodies, Tuebingen, TRP-1 protein vaccine, Colostrinin, anti-FAP antibody, YH-16, gemtuzumab, infliximab, cetuximab, trastuzumab, denileukin diftitox, rituximab, thymosin alpha 1 , bevacizumab, mecasermin, mecasermin rinfabate, oprelvekin, natalizumab, rhMBL, MFE-CP1 + ZD-2767-P, ABT-828, ErbB2-specific immunotoxin, SGN-35, MT-103, rinfabate, AS- 1402, B43-genistein, L-19 based radioimmunotherapeutics, AC-9301 , NY-ESO-1 vaccine, IMC-1 C11 , CT-322, rhCCI O, r(m)CRP, MORAb-009, aviscumine, MDX- 1307, Her-2 vaccine, APC-8024, NGR-hTNF, rhH1 .3, IGN-31 1 , Endostatin, volociximab, PRO- 1762, lexatumumab, SGN-40, pertuzumab, EMD-273063, L19-IL-2 fusion protein, PRX-321 , CNTO-328, MDX-214, tigapotide, CAT-3888, labetuzumab, alpha-particle-emitting radioisotope-llinked lintuzumab, EM- 1421 , HyperAcute vaccine, tucotuzumab celmoleukin, galiximab, HPV-16-E7, Javelin - prostate cancer. Javelin - melanoma, NY-ESO-1 vaccine, EGF vaccine, CYT-004-MelQbG10, WT1 peptide, oregovomab, ofatumumab, zalutumumab, cintredekin besudotox, WX-G250, Albuferon, aflibercept, denosumab, vaccine, CTP-37, efungumab, or 131 l-chTNT-1 /B. Monoclonal antibodies useful as the protein therapeutic include, but are not limited to, muromonab-CD3, abciximab, edrecolomab, daclizumab, gentuzumab, alemtuzumab, ibritumomab, cetuximab, bevicizumab, efalizumab, adalimumab, omalizumab, muromomab-CD3, rituximab, daclizumab, trastuzumab, palivizumab, basiliximab, and infliximab.
A compound of general formula (I ) as defined herein can optionally be administered in combination with one or more of the following: ARRY-1 62, ARRY-300, ARRY-704, AS- 703026, AZD-5363, AZD-8055, BEZ-235, BGT-226, B KM- 1 20, BYL-719, CAL- 101 , CC-223, CH- 51 32799, deforolimus, E-6201 , enzastaurin , GDC-0032, GDC-0068 , GDC-0623 , GDC-0941 , GDC-0973, GDC-0980, GSK-21 10183 , GSK-2126458, GSK-2141795, MK-2206, novolimus, OSI- 027, perifosine, PF-04691 502, PF-05212384, PX-866, rapamycin , RG-7167, RO-4987655, R0- 5126766, selumetinib, TAK-733 , trametinib, triciribine, UCN-01 , WX-554, XL- 147, XL-765, zotarolimus, ZSTK-474.
Generally, the use of cytotoxic and/or cytostatic agents in combination with a compound or composition of the present invention will serve to:
(1 ) yield better efficacy in reducing the growth of a tumor or even eliminate the tumor as compared to administration of either agent alone,
(2) provide for the administration of lesser amounts of the administered chemo- therapeutic agents,
(3 ) provide for a chemotherapeutic treatment that is well tolerated in the patient with fewer deleterious pharmacological complications than observed with single agent chemotherapies and certain other combined therapies,
(4) provide for treating a broader spectrum of different cancer types in mammals, especially humans,
(5) provide for a higher response rate among treated patients,
(6) provide for a longer survival time among treated patients compared to standard chemotherapy treatments,
(7) provide a longer time for tumor progression , and/or
(8) yield efficacy and tolerability results at least as good as those of the agents used alone, compared to known instances where other cancer agent combinations produce antagonistic effects. Methods of Sensitizing Cells to Radiation
In a distinct embodiment of the present invention, a compound of the present invention may be used to sensitize a cell to radiation. That is, treatment of a cell with a compound of the present invention prior to radiation treatment of the cell renders the cell more susceptible to DNA damage and cell death than the cell would be in the absence of any treatment with a compound of the invention. In one aspect, the cell is treated with at least one compound of the invention.
Thus, the present invention also provides a method of killing a cell, wherein a cell is administered one or more compounds of the invention in combination with conventional radiation therapy.
The present invention also provides a method of rendering a cell more susceptible to cell death, wherein the cell is treated with one or more compounds of the invention prior to the treatment of the cell to cause or induce cell death. In one aspect, after the cell is treated with one or more compounds of the invention, the cell is treated with at least one compound, or at least one method, or a combination thereof, in order to cause DNA damage for the purpose of inhibiting the function of the normal cell or killing the cell.
In one embodiment, a cell is killed by treating the cell with at least one DNA damaging agent. That is, after treating a cell with one or more compounds of the invention to sensitize the cell to cell death, the cell is treated with at least one DNA damaging agent to kill the cell. DNA damaging agents useful in the present invention include, but are not limited to, chemotherapeutic agents (e.g. , cisplatinum), ionizing radiation (X-rays, ultraviolet radiation), carcinogenic agents, and mutagenic agents.
In another embodiment, a cell is killed by treating the cell with at least one method to cause or induce DNA damage. Such methods include, but are not limited to, activation of a cell signalling pathway that results in DNA damage when the pathway is activated, inhibiting of a cell signalling pathway that results in DNA damage when the pathway is inhibited, and inducing a biochemical change in a cell, wherein the change results in DNA damage. By way of a non-limiting example, a DNA repair pathway in a cell can be inhibited, thereby preventing the repair of DNA damage and resulting in an abnormal accumulation of DNA damage in a cell.
In one aspect of the invention, a compound of the invention is administered to a cell prior to the radiation or other induction of DNA damage in the cell. In another aspect of the invention, a compound of the invention is administered to a cell concomitantly with the radiation or other induction of DNA damage in the cell. In yet another aspect of the invention, a compound of the invention is administered to a cell immediately after radiation or other induction of DNA damage in the cell has begun.
In another aspect, the cell is in vitro. In another embodiment, the cell is in vivo.
As mentioned supra, the compounds of the present invention have surprisingly been found to effectively inhibit tankyrases and may therefore be used for the treatment or prophylaxis of diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, or diseases which are accompanied with uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, particularly in which the uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses are affected by inhibition of tankyrases, such as, for example, haematological tumours, solid tumours, and/or metastases thereof, e.g. leukaemias and myelodysplastic syndrome, malignant lymphomas, head and neck tumours including brain tumours and brain metastases, tumours of the thorax including non-small cell and small cell lung tumours, gastrointestinal tumours, endocrine tumours, mammary and other gynaecological tumours, urological tumours including renal, bladder and prostate tumours, skin tumours, and sarcomas, and/or metastases thereof.
In accordance with another aspect therefore, the present invention covers a compound of general formula (I), or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, as described and defined herein, for use in the treatment or prophylaxis of a disease, as mentioned supra.
Another particular aspect of the present invention is therefore the use of a compound of general formula (I), described supra, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, for the prophylaxis or treatment of a disease.
Another particular aspect of the present invention is therefore the use of a compound of general formula (I) described supra or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, for manufacturing a pharmaceutical composition for the treatment or prophylaxis of a disease. Another aspect of the present invention is the use of a compound of formula (I) or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, as described herein, in the manufacture of a medicament for the treatment or prophylaxis of a disease.
The diseases referred to in the four preceding paragraphs are diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, or diseases which are accompanied with uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, such as, for example, haematological tumours, solid tumours, and/or metastases thereof, e.g. leukaemias and myelodysplastic syndrome, malignant lymphomas, head and neck tumours including brain tumours and brain metastases, tumours of the thorax including non-small cell and small cell lung tumours, gastrointestinal tumours, endocrine tumours, mammary and other gynaecological tumours, urological tumours including renal, bladder and prostate tumours, skin tumours, and sarcomas, and/or metastases thereof.
The term "inappropriate" within the context of the present invention, in particular in the context of "inappropriate cellular immune responses, or inappropriate cellular inflammatory responses", as used herein, is to be understood as meaning a response which is less than, or greater than normal, and which is associated with, responsible for, or results in, the pathology of said diseases.
Preferably, the use is in the treatment or prophylaxis of diseases, wherein the diseases are haemotological tumours, solid tumours and/or metastases thereof.
Diseases further included in the context of the present invention are metabolic diseases (e.g. diabetes and obesity), fibrosis (e.g. lung fibrogenesis) and viral infection. Method of treating hyper-proliferative disorders
The present invention relates to a method for using the compounds of the present invention and compositions thereof, to treat mammalian hyper-proliferative disorders. Compounds can be utilized to inhibit, block, reduce, decrease, etc. , cell proliferation and/or cell division, and/or produce apoptosis. This method comprises administering to a mammal in need thereof, including a human, an amount of a compound of this invention, or a pharmaceutically acceptable salt, isomer, polymorph, metabolite, hydrate, solvate or ester thereof ; etc. which is effective to treat the disorder. Hyperproliferative disorders include but are not limited, e.g. , psoriasis, keloids, and other hyperplasias affecting the skin, benign prostate hyperplasia (BPH), solid tumours, such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid and their distant metastases. Those disorders also include lymphomas, sarcomas, and leukaemias.
Examples of breast cancer include, but are not limited to invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ.
Examples of cancers of the respiratory tract include, but are not limited to small-cell and non-small-cell lung carcinoma, as well as bronchial adenoma and pleuropulmonary blastoma.
Examples of brain cancers include, but are not limited to brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal tumour.
Tumours of the male reproductive organs include, but are not limited to prostate and testicular cancer. Tumours of the female reproductive organs include, but are not limited to endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus.
Tumours of the digestive tract include, but are not limited to anal, colon, colorectal, oesophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers.
Tumours of the urinary tract include, but are not limited to bladder, penile, kidney, renal pelvis, ureter, urethral and human papillary renal cancers.
Eye cancers include, but are not limited to intraocular melanoma and retinoblastoma. Examples of liver cancers include, but are not limited to hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma.
Skin cancers include, but are not limited to squamous cell carcinoma, Kaposi 's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer.
Head-and-neck cancers include, but are not limited to laryngeal, hypopharyngeal, nasopharyngeal, oropharyngeal cancer, lip and oral cavity cancer and squamous cell. Lymphomas include, but are not limited to AlDS-related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Burkitt lymphoma, Hodgkin's disease, and lymphoma of the central nervous system.
Sarcomas include, but are not limited to sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma.
Leukemias include, but are not limited to acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia.
These disorders have been well characterized in humans, but also exist with a similar etiology in other mammals, and can be treated by administering pharmaceutical compositions of the present invention.
The term "treating" or "treatment" as stated throughout this document is used conventionally, e.g. , the management or care of a subject for the purpose of combating, alleviating, reducing, relieving, improving the condition of, etc. , of a disease or disorder, such as a carcinoma.
Methods of treating angiogenic disorders
The present invention also provides methods of treating disorders and diseases associated with excessive and/or abnormal angiogenesis.
Inappropriate and ectopic expression of angiogenesis can be deleterious to an organism. A number of pathological conditions are associated with the growth of extraneous blood vessels. These include, e.g. , diabetic retinopathy, ischemic retinal-vein occlusion, and retinopathy of prematurity [Aiello et al. New Engl. J. Med. 1994, 331 , 1 80 ; Peer et al. Lab. Invest. 1995, 72, 638], age-related macular degeneration [AMD ; see, Lopez et al. Invest. Opththalmol. Vis. Sci. 1996, 37, 855] , neovascular glaucoma, psoriasis, retrolental fibroplasias, angiofibroma, inflammation, rheumatoid arthritis (RA), restenosis, in-stent restenosis, vascular graft restenosis, etc. In addition, the increased blood supply associated with cancerous and neoplastic tissue, encourages growth, leading to rapid tumour enlargement and metastasis. Moreover, the growth of new blood and lymph vessels in a tumour provides an escape route for renegade cells, encouraging metastasis and the consequence spread of the cancer. Thus, compounds of the present invention can be utilized to treat and/or prevent any of the aforementioned angiogenesis disorders, e.g. , by inhibiting and/or reducing blood vessel formation ; by inhibiting, blocking, reducing, decreasing, etc. endothelial cell proliferation or other types involved in angiogenesis, as well as causing cell death or apoptosis of such cell types.
Dose and administration
Based upon standard laboratory techniques known to evaluate compounds useful for the treatment of hyper- proliferative disorders and angiogenic disorders, by standard toxicity tests and by standard pharmacological assays for the determination of treatment of the conditions identified above in mammals, and by comparison of these results with the results of known medicaments that are used to treat these conditions, the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication. The amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
The total amount of the active ingredient to be administered will generally range from about 0.001 mg/ kg to about 200 mg/kg body weight per day, and preferably from about 0.01 mg/kg to about 20 mg/ kg body weight per day. Clinically useful dosing schedules will range from one to three times a day dosing to once every four weeks dosing. In addition, "drug holidays" in which a patient is not dosed with a drug for a certain period of time, may be beneficial to the overall balance between pharmacological effect and tolerability. A unit dosage may contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered one or more times per day or less than once a day. The average daily dosage for administration by injection, including intravenous, intramuscular, subcutaneous and parenteral injections, and use of infusion techniques will preferably be from 0.01 to 200 mg/ kg of total body weight. The average daily rectal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily vaginal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily topical dosage regimen will preferably be from 0.1 to 200 mg administered between one to four times daily. The transdermal concentration will preferably be that required to maintain a daily dose of from 0.01 to 200 mg/kg. The average daily inhalation dosage regimen will preferably be from 0.01 to 100 mg/kg of total body weight.
Of course the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like. The desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
Preferably, the diseases of said method are haematological tumours, solid tumour and/or metastases thereof.
The compounds of the present invention can be used in particular in therapy and prevention, i.e. prophylaxis, of tumour growth and metastases, especially in solid tumours of all indications and stages with or without pre-treatment of the tumour growth.
Methods of testing for a particular pharmacological or pharmaceutical property are well known to persons skilled in the art.
The example testing experiments described herein serve to illustrate the present invention and the invention is not limited to the examples given.
Biological assays:
Examples were tested in selected biological assays one or more times. When tested more than once, data are reported as either average values or as median values, wherein
• the average value, also referred to as the arithmetic mean value, represents the sum of the values obtained divided by the number of times tested, and • the median value represents the middle number of the group of values when ranked in ascending or descending order. If the number of values in the data set is odd, the median is the middle value. If the number of values in the data set is even, the median is the arithmetic mean of the two middle values.
Examples were synthesized one or more times. When synthesized more than once, data from biological assays represent average values or median values calculated utilizing data sets obtained from testing of one or more synthetic batch.
6 Biochemical assays
6.1 TNKS1 Assays
TNKS1 Assay A
The potency of the compounds according to the invention was assessed by applying an in vitro inhibition assay. The TNKS1 catalyzed NAD'-dependent ribosylation of a suitable protein substrate was detected using a commercially available biotin/streptavidin binding based assay format [TNKS1 Histone Ribosylation Assay Kit (Biotin-labeled NAD+), Catalog #80579; BPS Bioscience, San Diego, USA] . Here, the incorporation of a biotin-labeled NAD+ during the TNKS1 catalyzed ribosylation reaction was detected with a streptavidin-HRP coupled chemi-luminescent readout. The intensity of the readout signal is proportional to the incorporated NAD'. Inhibition of TNKS1 leads to a decreased incorporation of NAD* and consequently to a lower readout signal intensity. The concentration of a test compound which inhibits the enzyme activity by 50% (corresponds to half of the normed readout signal intensity) is reported as ICw.
Protocol
The assay was conducted in a 384 well MTP format according to the manufacturer's protocol [http: / /www.bpsbioscience.com/poly-adp-ribose-polymerase/assay-kit/tnks1 - histone-ribosylation-assay-kit-biotin-labeled-nad-80579 referencing: Brown, J. A. , Marala, R. B. J. Pharmacol. Toxicol. Methods 2002 47: 137] and using a BMG Pherastar MTP reader [BMG-Labtech, Offenburg, Germany] .
TNKS1 Assay B The potency of selected compounds according to the invention was assessed applying a modified in vitro inhibition assay. Here, the TNKS1 catalyzed NAD+-dependent ribosylation of the enzyme itself (auto-parsylation) was detected using [ H]-NAD* as substrate and applying the scintillation proximity assay (SPA) method to detect tritium-labeled, parsylated TNKS1 . The intensity of the readout signal is proportional to the incorporated [3H]-NAD*. Inhibition of TNKS1 leads to a decreased incorporation of [3H]-NAD+ and consequently to a lower readout signal intensity. The concentration of a test compound which inhibits the enzyme activity by 50% (corresponds to half of the normed readout signal intensity) is reported as IC».
Protocol Auto-Parsylation Assay
The assay was conducted in a 96 well MTP format with the identical TNKS1 enzyme sample and NAD* sample as in the histone ribosylation assay with the following modifications: TNKS1 enzyme sample was diluted with a modified assay buffer (50 mM MES pH 7.0, 1 mM DTT, 0.01 % Triton X-100) to a final concentration of 6 nM TNKS1 and 10x NAD* solution was diluted with the modified assay buffer (s. above) to a final 0.445x NAD' solution doped with 100 Bq/μΙ [3H]-NAD+ [Catalog #NET443H050UC, Perkin Elmer, Waltham , Massachusetts, USA ]. Substrate solution (10 μΐ) was incubated with different test compound concentrations (2.5 ul in 10 % DMSO in modified assay buffer) or control (2.5 ul 10 % DMSO in modified assay buffer only) and enzyme (10 μΐ) over night at room temperature. Incorporated tritium was measured after addition of 50 μΐ SPA beads (1 mg/ml) [Catalog #RPNQ0095 20 mg/ ml, Perkin Elmer, Waltham, Massachusetts, USA; diluted 1 : 10 with Dulbecco's phosphate buffered saline, PBS Catalog #08537, Sigma- Aldrich, Steinheim, Germany] and detection of the photon emission with a beta count plate reader [Wallac MicroBeta®, Perkin Elmer, Waltham , Massachusetts, USA]. 6.2 TNKS2 Assays
TNKS2 Assay A
The potency of the compounds according to the invention was assessed applying an in vitro inhibition assay. The TNKS2 catalyzed NAD'-dependent ribosylation of a suitable protein substrate was detected using a commercially available biotin/streptavidin binding based assay format [TNKS2 Histone Ribosylation Assay Kit (Biotin-labeled NAD'), Catalog #80572; BPS Bioscience, San Diego, USA]. Here, the incorporation of a biotin-labeled NAD* during the TNKS2 catalyzed ribosylation reaction was detected with a streptavidin-HRP coupled chemi-luminescent readout. The intensity of the readout signal is proportional to the incorporated NAD*. Inhibition of TNKS2 leads to a decreased incorporation of NAD* and consequently to a lower readout signal intensity. The concentration of a test compound which inhibits the enzyme activity by 50% (corresponds to half of the normed readout signal intensity) is reported as IC¾o.
Protocol
The assay was conducted in a 384 well MTP format according to the manufacturer's protocol [http: / /www. bpsbioscience.com/ poly-adp-ribose-polymerase/assay-kit/tnks2- histone-ribosylation-assay-kit-biotin-labeled-nad-80572 referencing: Brown, J. A. , Marala, R. B. J. Pharmacol. Toxicol. Methods 2002 47: 137] . and using a BMG Pherastar MTP reader [BMG-Labtech, Offenburg, Germany] .
TNKS2 Assay B
The potency of selected compounds according to the invention was assessed applying a modified in vitro inhibition assay. Here, the TNKS2 catalyzed NAD'-dependent ribosylation of the enzyme itself (auto-parsylation ) was detected using [3H] -NAD* as substrate and applying the scintillation proximity assay (SPA) method to detect tritium-labeled, parsylated TNKS2. The intensity of the readout signal is proportional to the incorporated [3H] -NAD\ I nhibition of TNKS2 leads to a decreased incorporation of [3H]-NAD* and consequently to a lower readout signal intensity. The concentration of a test compound which inhibits the enzyme activity by 50% (corresponds to half of the normed readout signal intensity) is reported as IC».
Protocol Auto-Parsylation Assay
The assay was conducted in a 96 well MTP format with the identical TNKS2 enzyme sample and NAD' sample as in the histone ribosylation assay with the following modifications: TNKS2 enzyme sample was diluted with a modified assay buffer (50 mM MES pH 7.0, 1 mM DTT, 0.01 % Triton X-100) to a final concentration of 6 nM TNKS2 and 10x NAD' solution was diluted with the modified assay buffer (s. above) to a final 0.445x NAD* solution doped with 100 Bq/ μΙ [3H] -NAD+ [Catalog #NET443H050UC, Perkin Elmer, Waltham , Massachusetts, USA] . Substrate solution (10 μΐ) was incubated with different test compound concentrations (2.5 μΐ in 10 % DMS0 in modified assay buffer) or control (2.5 μΐ 10 % DMS0 in modified assay buffer only) and enzyme (10 μΐ) over night at room temperature. Incorporated tritium was measured after addition of 50 ul SPA beads (1 mg/ ml) [Catalog #RPNQ0095 20 mg/ ml, Perkin Elmer, Waltham , Massachusetts, USA; diluted 1 : 10 with Dulbecco's phosphate buffered saline, PBS Catalog #08537, Sigma- Aldrich, Steinheim, Germany] and detection of the photon emission with a beta count plate reader [Wallac Micro Beta®, Perkin Elmer, Waltham , Massachusetts, USA].
6.3 PARP 1 Assay
The potency of the compounds according to the invention was assessed using a commercially available biotin/streptavidin binding assay kits from BPS Bioscience, San Diego, USA (Catalog #80551 ). The incorporation of a biotin-labeled NAD' during the PARP1 catalyzed ribosylation of a suitable protein substrate was detected using with a streptavidin-HRP coupled chemi-luminescent readout. The intensity of the readout signal is proportional to the incorporated NAD*. Inhibition of PARP1 leads to a decreased incorporation of NAD' and consequently to a lower readout signal intensity. The concentration of a test compound that inhibits the enzyme activity by 50% (corresponds to half of the normed readout signal intensity) is reported as ICso-
Protocol
The assay was conducted in a 96 well MTP format according to the manufacturer's protocol (Catalog No. 80551 ) and using a BMG Pherastar MTP reader [BMG-Labtech, Offenburg, Germany].
6.4 PARP2 Assay
The potency of the compounds according to the invention was assessed using a commercially available biotin/streptavidin binding assay kits from BPS Bioscience, San Diego, USA (Catalog #80551 ). The incorporation of a biotin-labeled NAD* during the PARP2 catalyzed ribosylation of a suitable protein substrate was detected using with a streptavidin-HRP coupled chemi-luminescent readout. The intensity of the readout signal is proportional to the incorporated NAD'. Inhibition of PARP2 leads to a decreased incorporation of NAD* and consequently to a lower readout signal intensity. The concentration of a test compound that inhibits the enzyme activity by 50% (corresponds to half of the normed readout signal intensity) is reported as ICso.
Protocol
The assay was conducted in a 96 well MTP format according to the manufacturer's protocol (Catalog No. 80552) and using a BMG Pherastar MTP reader [BMG-Labtech, Offenburg, Germany]. 7 Cellular Assays
7.1 Measurement of the inhibitory activity of selected compounds on the Wildtype Wnt signaling cascade: HEK293 TOP/FOP Assay
In order to discover and characterize small molecules which inhibit the wildtype Wnt pathway, a cellular reporter assay was employed. The corresponding assay cell was generated by transfection of the mammalian cell line HEK293 (ATCC, #CRL-1 73) with the Super TopFlash vector (Morin, Science 275, 1997, 1787-1790; Molenaar et al., Cell 86 (3), 1996, 391 -399). The HEK293 cell line is cultivated at 37oC and 5% C02 in DMEM (Life Technologies, #41965-039), supplemented with 2 m glutamine, 20 mM HEPES, 1.4 mM pyruvate, 0.15% Na-bicarbonate and 10% foetal bovine serum (GIBCO, #10270). Stable transfectants were generated by selection with 300 g/ml Hygromycin.
In a parallel approach, HEK293 cells were cotransfected with the FOP control vector and pcDNA3. The FOP vector is identical to the TOP construct, but it contains instead of functional TCF elements a randomized, non-functional sequence. For this transfection a stable transfected cell line was generated as well, based on selection with Geneticin (1 mg/ml).
In preparation of the assay, the two cell lines were plated 24 h before beginning the test at 10000 cells per well in a 384 micro tit re plate (MTP) in 30 μΐ growth medium. Before compound testing a dose response curve for the Wnt dependent luciferase expression was recorded by stimulating the assay cell line with human recombinant Wnt-3a (R&D, #5036- WN-010) at different concentrations for 16 h at 37 C and 5% CO2 followed by subsequent luciferase measurement, to determine the Wnt-3a EC50 for the HEK293 TOP cell line on the day of testing. The recombinant human Wnt-3a was thereby applied between 2500 and 5 ng/ml in two-fold dilution steps.
Selective inhibitory activity for small molecules on the wildtype Wnt pathway was determined after parallel incubation of both (TOP and FOP) HEK293 reporter cell lines with a compound dilution series from 50 μΜ to 15 nM in steps of 3.16-fold dilutions in CAFTY buffer (130 mM sodium chloride, 5 mM potassium chloride, 20 mM HEPES, 1 mM magnesium chloride, 5 mM sodium bicarbonate, pH 7.4) containing 2 mM Ca2+ and 0.01% BSA.
The compounds were thereby serially prediluted in 100% DMSO and thereafter 50 fold into the CAFTY compound dilution buffer (described above). From this dilution 10 μΐ were added in combination with the EC 50 concentration of recombinant Wnt3a to the cells in 30 μΐ growth medium and incubated for 16 hours at 37 C and 5% CO?. Thereafter luciferase assay buffer (1 : 1 mixture of luciferase substrate buffer (20 mM Tricine, 2.67 mM magnesium sulfate, 0.1 mM EDTA, 4 mM DTT, 270 μΜ Coenzyme A, 470 μΜ Luciferin, 530 μΜ ATP, ph adjusted to pH 7.8 with a sufficient volume of 5M sodium hydroxide) and Triton buffer (30 ml Triton X-100, 1 15 ml glycerol, 308 mg Dithiothreitol, 4.45 g disodium hydrogen phosphate di hydrate, 3.03 g Tris . HQ, ad 11 H?0, pH 7.8) was added in an equal volume to determine luciferase expression as a measure of Wnt signaling activity in a luminometer. The Wnt inhibitory activity was determined as IC50 of resulting dose response curves.
7.2 Axin Stabilization Assay
The in vitro and in vivo effect of Tankyrase inhibition on the stabilization of cellular Axin was assessed using Peggy Simple Western assay with size-based separation and immunodetection of Axin2. SW403 cells (but not limited to) were seeded at 50000 cells per well in 96-well plates. After overnight incubation, cells were treated with testing compounds and vehicle at 37 C for 24 hours. Thereafter, cells were washed with PBS and then lysed in 15 μΐ of lysis buffer (M-PER buffer, Thermo Scientific # 78505) with complete proteinase and phosphatase inhibitors (Roche, #11836153001 and # 04906837001 ). The lysates were centrifuged and the supernatants were harvested for analysis. Tumor xenografts from in vivo studies were homogenized in a 2 ml tubes of Precellysl24 (Bertin Technologies, Villeurbanne, France) following with centrafugation to obtain tumor lysates. Capillary electrophoresis-based Simple Western assays were carried out with Peggy Sue™ NanoPro 1000 (ProteinSimple, California, USA). The protein amounts of Axin2 (but not limited to) were detected using anti-Axin2 antibody (Cell Signaling, Catalog #2151 ), quantified using the area under the curve, and normalized against GAPDH (anti-GAPDH, Zytomed Systems GmbH, Catalog #RGM2-6C5, Berlin, Germany).
7.3 Real-time RT-PCR for quantitative analysis of gene transcription
Real-time RT-PCR using a TaqMan fluorogenic detection system is a simple and sensitive assay for quantitative analysis of gene transcription. The TaqMan fluorogenic detection system can monitor PCR in real time using a dual-labeled fluorogenic hybridization probe (TaqMan probe) and a polymerase with 5'-3' exonuclease activity. Cells from different cancer cell lines (as HCT116, but not limited to) were grown at 500- 1000 cells/well in 384 well cell culture plates. For cell lysis the cell medium was carefully removed. The cells were washed carefully once with 50 μΐ/well PBS. Then 9.75 μΐ/well cell lysis buffer (50 tri Tris HCl pH 8,0, 40 mM sodium chloride, 1 ,5 mM magnesium chloride, 0,5 % IGEPAL CA 630, 50mM Guanidium thiocyanate) and 0.25 μΐ RNASeOUT (40 U/μΙ, Invitrogen, 10777-019)) per well were added. The plate was incubated for 5 min at room temperature. Then 30 μΐ DNAse/ RNAse-f ree water per well was added and the lysates mixed. Isolation of total RNA from tumor tissues was conducted using InviTrap® Spin Tissue RNA Mini Kit (#1062100300, STRATEC MOLECULAR).
For the One-Step RT-PCR 2 μΐ lysate (each) was transferred to a 384 well PCR plate. The PCR reaction was composed by 5 μΐ 2x One Step RT qPCR MasterMix Plus, 0.05 μΐ Euroscript RT/RNAse Inhibitor (50 U/μΙ, 20 SJ/μ ) and 200 nM of the appropriate Primer/ Hydrolysis Probe mix (primer sequences of forward, reverse and probe are given below for each analysed gene of interest or house keeping gene). 10 μΐ water were added per well. The plate was sealed with an adhesive optical film. The RT-PCR protocol was setup with 30 min 48 C, then 10 min 95 C followed by 50 cycles of 15 sec 95 C/1 min 60 C and a cooling step of 40 C for 30 sec using a Lightcycler LS440 from Roche. Relative expression was calculated using CP values from the gene of interest (e.g. AXIN2, but not limited to) and a house keeping gene (L32).
Used primers
L32 (forward primer: AAGTTCATCCGGCACCAGTC (SEQ. ID NO. 1 ); reverse primer: TGGCCCTTGAATCTTCTACGA (SEQ ID NO. 2); probe: CCCAGAGGCATTGACAACAGGG (SEQ ID NO. 3))
AXIN2 (forward primer: AGG CCAGTG AGTTG GTTGTC (SEQ ID NO. 4); reverse primer: AGCTCTGAGCCTTCAGCATC (SEQ ID NO. 5); probe: TCTGTGGGGAAGAAATTCCATACCG (SEQ ID NO. 6))
8 In vivo Efficacy in xenograft models
Subcutaneous xenograft models in immunocompromised mice were used to evaluate in vivo anti-tumor efficacy of the compounds. 8. 1 Maximum tolerable dose (MTD) studies
Prior to efficacy studies, the maximal tolerable dose (MTD) was determined by the following protocol: Female nude mice (NMRI (nu/ nu), Taconic M&B A/ S) received a defined oral dose of the test compound daily or bi-daily for 7 consecutive days followed by a 7 day observation period without dosing. Individual body weight and lethality were monitored daily.
The MTD is defined as the maximal applicable dose with a) no animal losing more than 10% body weight compared to initial body weight and b) no lethality during treatment phase.
8.2 In vivo efficacy studies
To measure anti-tumor efficacy, the test compounds were analysed in xenograft models on mice. Test compounds were dosed orally at their respective MTD as well as at sub-MTD dosages. In case the MTD could not be determined in previous MTD studies, the compounds were dosed at a maximum daily dose of 200 mg/kg (applied either in one single dose or split in 2 doses at 100 mg/ kg).
Compounds were primarily analyzed in an ovarian teratocarcinoma model (PA-1 ) and in various colorectal cancer models on female immunocompromised mice.
For this purpose, 1 -5x10* tumor cells (suspended in 0.1 ml of 50% cell culture medium / 50% Matrigel) were subcutanously injected into the flank of each animal. Animals were randomized into treatment groups when tumors had reached an average area of 20-30 mm2 and treatment was started. Body weight and tumor area of each animal were measured 2-3 times weekly, depending on tumor growth. Studies were terminated , when animals in the control groups (receiving only compound vehicle solutions) or treatment groups reached tumor areas - 1 50 mm2. At that time point, all groups in the study were terminated, tumors were isolated and weighed.
As primary parameter for anti-tumor efficacy the Treatment/Control (T/C) ratio of the final tumor weights were calculated (mean tumor weight of treatment group divided by mean tumor weight of vehicle group). 8.3 In vivo Mode of Action studies
To determine in vivo Mode of Action (MoA) of the test compounds, the same in vivo models as described under 8.2 were utilized. Tumor-bearing animals were treated for at least 3 days at MTD and also sub-MTD dosages. At study end, tumors were isolated and snap frozen in liquid nitrogen. Total RNA and protein were isolated from tumor samples following standard protocols.
Wnt/B-catenin target gene expression and Axin2 protein abundance were measured by standard qRT-PCR and Western blotting methods (see 7.2 and 7.3).
Table 1 : IC¾o values for selected examples in cellular HEK293 TOP and FOP assay as well as in TNKS1 and TNKS2 biochemical assay
Figure imgf000115_0001
5.3 50 > 10
2.3 10 0.82
0.91 27 0.36
0.14 7.2 0.0068
0.11 50 0.16
0.37 6.2 0.011
0.26 6.6 0.27

Claims

1 . A compound of formula (I)
Figure imgf000117_0001
in which :
X1 represents NR3 or 0.
X2 represents CR6 or N ,
R1 represents a group selected from :
-OR9, and -N(R,0)R" ,
R2 represents a group selected from :
hydrogen , O -Cralkyl, and CrC-cycloalkyl,
R3 represents hydrogen ,
R4 represents hydrogen ,
R5 represents a group selected from :
hydrogen , and Ci -Cs-alkyl,
R6 represents a group selected from :
hydrogen, and halogen ,
R7 represents hydrogen ,
R8 represents a group selected from :
aryl, aryl-(CrG.-alkyl)-, heteroaryl, and heteroaryl- (CrC,-alkyl)- ,
wherein aryl and heteroaryl groups are optionally substituted with one, two or three substituents, which are independently of each other selected from : G-G-alky , G-Gralkoxy, G-Grhydroxyalkyl, GrG-cycloalkyl, G-G-cycloalkoxy, G-Grhaloalkyl, G-G-haloalkoxy, halogen, cyano, nitro, hydroxy, -N(R'°)R" , R'°(R" )N-(G-G.-alkyl)-, R10(R11 )N-(C2-C4-alkoxy)-,
R9 represents G-Gralkyl,
R10 and R" are independently of each other selected from :
hydrogen, G -Cralkyl, G-G-cycloalkyl, (G-G-cycloalkyl)-(G-G-alkyl)-,
G-G-hydroxyalkyl, (G-G-alkoxy)-(GrG-alkyl)-, G-G-haloalkyl,
H2N-(G-G-alkyl)-, (G-G-alkyl)N(H )(G-G-alkyl)-, (G -G-alkylbN(G-G-alkyl)-, 4-6 membered heterocycloalkyl, (4- to 6-membered heterocycloalkyl)- (G-G-alkyl)-, wherein 4- to 6-membered heterocycloalkyl groups are optionally substituted with one or two substituents, which are independently of each other selected from :
G-alkyl, G-haloalkyl, G -alkoxy, G -hydroxyalkyl, G-haloalkoxy, halogen, and hydroxy; or,
R'° and R" together with the nitrogen atom to which they are attached form a 4- to 6-membered heterocycloalkyl group, in which one carbon atom is optionally replaced by a further heteroatom-containing group selected from NR12, and 0, in which heterocycloalkyl group one additional ring atom is optionally replaced by C(=0), said 4-to 6-membered heterocycloalkyl group being optionally substituted with one or two substituents, which are independently of each other selected from :
G-alkyl, G-haloalkyl, G -alkoxy, G -haloalkoxy, halogen, and hydroxy;
R12 represents a group selected from :
hydrogen, and G-alkyl,
or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.
2. The compound according to claim 1 , wherein :
X1 represents NR3,
X2 represents CR6,
R1 represents a group selected from : -OR9, and -N(R10)R11 ,
R2 represents a group selected from :
hydrogen, and Ci-alkyl,
R3 represents hydrogen,
R4 represents hydrogen,
R5 represents a group selected from :
hydrogen, and Ci-alkyl,
R6 represents a group selected from :
hydrogen, and fluorine,
R7 represents hydrogen,
R8 represents a group selected from :
aryl, aryl-(Ci-C4-alkyl)-, heteroaryl, and heteroaryl-(Ci-C4-alkyl)-,
wherein aryl and heteroaryl groups are optionally substituted with one, two or three substituents, which are independently of each other selected from :
Ci-Ci-alkyl, d-Cs-alkoxy, d-d-hydroxyalkyl, d-drhaloalkyl, halogen, cyano, -N(R'°)R",
R9 represents Ci-Cz-alkyl,
R'° and R11 are independently of each other selected from :
hydrogen, Ci-Ci-alkyl, rG-cycloalkyl, (d-d-cycloalkylHCi-Ci-alkyl)-,
Crd-hydroxyalkyl, (Ci-alkoxy)-(C?-C3-alkyl)-, Ci-Crhaloalkyl, hhN-iCrd-alkyl)-, (Ci- alkyl)N(H)(C2-C3-alkyl)-, (C,-alkyl N(Crdralkyl)-, (4-6 membered heterocycloalkyl)- (C C3- alkyl)-, wherein 4-6-membered heterocycloalkyl groups are optionally substituted with one or two substituents, which are independently of each other selected from :
Ci-alkyl, Ci-haloalkyl, Ci-alkoxy, d-hydroxyalkyl, Ci-haloalkoxy, halogen, and hydroxy; or,
R'° and R" together with the nitrogen atom to which they are attached form a
4-6-membered heterocycloalkyl group, in which one carbon atom is optionally replaced by a further heteroatom-containing group selected from NR", and 0, in which heterocycloalkyl group one additional ring atom is optionally replaced by C(=0), said 4-6-membered heterocycloalkyl group being optionally substituted with one or two substituents, which are independently of each other selected from :
G-alkyl, Ci-haloalkyl, Ci-alkoxy, d-haloalkoxy, halogen, and hydroxy;
R12 represents a group selected from :
hydrogen, and G-alkyl,
or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof , or a mixture of same.
3. The compound according to any one of claims 1 and 2, wherein :
X1 represents NR3,
X2 represents CR6,
R1 represents -N(R'°)R",
R2 represents hydrogen,
R3 represents hydrogen,
R4 represents hydrogen,
R5 represents hydrogen,
R6 represents hydrogen,
R7 represents hydrogen,
R8 represents a group selected from
aryl, aryl-(G-Cralkyl)-,
wherein aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
halogen,
R'° and R11 are independently of each other selected from :
hydrogen, G-G-alkyl, G-G-cycloalkyl, (G-G-cycloalkyl)-(G-alkyl)-,
CrG-hydroxyalkyl, (G-alkoxy)-(C?-G-alkyl)-, G-Crhaloalkyl, HiN-(Cralkyl)-, (G- alkyl)N(H)(G-alkyl)-, (G-alkyl )2N(C alkyl)-, (6-membered heterocycloalkyl)-(C2-alkyl)-, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.
4. The compound according to any one of claims 1 , 2 or 3, wherein :
X1 represents NR3,
X2 represents CR6,
R' represents -N(R! 0)R" ,
R2 represents hydrogen,
R3 represents hydrogen,
R4 represents hydrogen,
R5 represents hydrogen,
R6 represents hydrogen,
R7 represents hydrogen,
R8 represents a group selected from
aryl, aryl-(CrCralkyl)-,
wherein aryl is optionally substituted with one, two or three substituents, which are independently of each other selected from :
fluorine and chlorine,
R'° and R11 are independently of each other selected from :
hydrogen, C C2-alkyl, (C, -alkyl)2N(C2-alkyl)-, (piperidin-1 -yl)-(C2-alkyl)-.
or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.
5. The compound according to any one of claims 1 to 4, which is selected from the group consisting of :
N5-{4-[(3-chlorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4,5- dicarboxamide,
N5-{4-[(3-chlorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4,5- dicarboxamide formic acid salt, N5-{4-[(2-chloro-4-fluorobenzyl)carbamoyl]phenyl}-N -[2-(piperidin-1 -yl)ethyl]-1 H- imidazole-4, 5-dicarboxamide,
N5-{4-[(2-chloro-4-fluorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin-1 -yl)ethyl]-1 H- imidazole-4, 5-dicarboxamide formic acid salt,
N5-{4-[(3, 5-dichlorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole- 4,5-dicarboxamide,
N5-{4-[(3, 5-dichlorobenzyl)carbamoyl]phenyl}-N4-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole- 4, 5-dicarboxamide formic acid salt,
N4-{4-[(3, 5-dichlorophenyl)carbamoyl]phenyl}-N5-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole- 4, 5-dicarboxamide,
N4-{4-[(3, 5-dichlorophenyl)carbamoyl]phenyl}-N5-methyl-1 H-imidazole-4,5- dicarboxamide,
N4-{4- [(2, 3-dichlorophenyl)carbamoyl]phenyl}-N5- [2- (piperidin-1 -yl)ethyl]-1 H-imidazole- 4,5-dicarboxamide,
N4-{4-[(3-chlorophenyl)carbamoyl]phenyl}-N5-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4, 5- dicarboxamide,
N -{4-[(3-chlorophenyl)carbamoyl]phenyl}-N5-methyl-1 H-imidazole-4, 5-dicarboxamide,
N -{4-[(2, 3-dichlorophenyl)carbamoyl]phenyl}-N5-methyl-1 H-imidazole-4,5- dicarboxamide,
N4-(4-{[1 -(2-chlorophenyl)ethyl]carbamoyl}phenyl)-N5-[2-(piperidin-1 -yl)ethyl]-1 H- imidazole-4, 5-dicarboxamide,
N4-(4-{[1 -(2-chlorophenyl)ethyl]carbamoyl}phenyl)-N5-methyl-1 H-imidazole-4,5- dicarboxamide,
N -{4-[(2, 3-dichlorobenzyl)carbamoyl]phenyl}-N5-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole- 4, 5-dicarboxamide,
N4-{4-[(2, 3-dichlorobenzyl)carbamoyl]phenyl}-N5-methyl-1 H-imidazole-4,5- dicarboxamide,
N5-{4-[(2-chlorophenyl)carbamoyl]phenyl}-N4-methyl-1 H-imidazole-4, 5-dicarboxamide,
N5-{4-[(2-chloro-4-fluorophenyl)carbamoyl]phenyl}-N4-methyl-1 H-imidazole-4,5- dicarboxamide, N5-{4-[(4-fluorophenyl)carbamoyl]phenyl}-N4-methyl-1 H-imida
N4- [2- (di methylamino)ethyl] - N5-{4- [(4-fluorophenyl)carbamoyl] phenyl}- 1 H-imidazole- 4,5-dicarboxamide,
N5-{4-[(4-fluorophenyl)carbamoyl]phenyl}-N4-[2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4, 5- dicarboxamide,
N¾-{4-[(2-chloro-4-fluorophenyl)carbamoyl]phenyl}-N4-[2-(piperidin-1 -yl)ethyl]-1 H- imidazole-4,5-dicarboxamide,
N5-{4-[(2-chloro-4-fluorophenyl)carbamoyl]phenyl}-N4-[2-(dimethylamino)ethyl]-1 H- imidazole-4,5-dicarboxamide,
N5-{4-[(2-chlorophenyl)carbamoyl]phenyl}-N -[2-(piperidin-1 -yl)ethyl]-1 H-imidazole-4.5- dicarboxamide, and
N¾-{4-[(2-chlorophenyl)carbamoyl]phenyl}-N4-[2-(dimethylamino)ethyl]-1 H-imidazole-4, 5- dicarboxamide,
or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.
6. A method of preparing a compound of general formula (I) according to any one of claims 1 to 5, said method comprising the step of allowing an intermediate compound of general formula (III) :
Figure imgf000123_0001
(Hi)
in which Xi represents N, and R! , and R2 are as defined for the compound of general formula (I) in any one of claims 1 to 5, to react with a compound of general formula (II)
Figure imgf000124_0001
in which X2, R4, R5, R7, and R8 are as defined for the compound of general formula (I ) according to any one of claims 1 to 5,
thereby giving a compound of general formula (!) :
Figure imgf000124_0002
(I) in which Xt represents NR3, and X2, R\ R2, R3, R5, R7, and R8 are as defined for the compound of general formula (I) in any one of claims 1 to 5.
7. A method of preparing a compound of general formula (I ) according to any one of claims 1 to 5, said method comprising the step of allowing an intermediate compound of general formula (3-4) :
Figure imgf000124_0003
in which Xi represents NR3. X2, R1 , R2, R3, R4, R5, and R9 are as defined for the compound of general formula (I ) in any one of claims 1 to 5 and R13 represents R9 or H . to react with a compound of general formula (1 -2) :
Figure imgf000125_0001
1-2
in which R7, and R8 are as defined for the compound of general formula (I) in any one of claims 1 to 5, thereby giving a compound of general formula (I)
Figure imgf000125_0002
(I) in which X, represents NR3, and X2, R\ R2, R4, R5, R7, and R8 are as defined for the compound of general formula (I) in any one of claims 1 to 5.
8. A method of preparing a compound of general formula (I) according to any one of claims 1 to 5, said method comprising the step of allowing an intermediate compound of general formula (VI ) :
Figure imgf000125_0003
in which Xi represents N 3, X2, R2, R3, R4, R5, R7 and R8 are as defined for the compound of general formula (I ) in any one of claims 1 to 5,
to react with a compound of general formula (1 -2) :
R"-H
3-2 in which R1 represents represents N(R10)R11 and R10, and R11 are as defined for the compound of general formula (I) in any one of claims 1 to 5,
thereby giving a compound of general formula (I ) :
Figure imgf000126_0001
(I)
ί
in which X1 represents NR3, R1 represents N(R10)R11 and X2, R2, R\, R4, R5, R7, R8, R'°, and R11 are as defined for the compound of general formula (I) in any one of claims 1 to 5.
9. A method of preparing a compound of general formula (I ) according to any one of claims 1 to 5, said method comprising the step of allowing an intermediate compound of general formula (IV) :
Figure imgf000126_0002
in which X1 represents N , and X2, R2, R4, R5, R7 and R8 are as defined for the compound of general formula (I) in any one of claims 1 to 5,
to react with a compound of general formula (1 -2) :
R1-H
3-2
in which R1 is as defined for the compound of general formula (I ) in any one of claims 1 to
5,
thereby giving a compound of general formula (I )
Figure imgf000127_0001
(I) in which X1 represents NR3 and X2, R\, R2, R3, R4, R5, R7, and R8 are as defined for the compound of general formula (I) in any one of claims 1 to 5.
10. A method of preparing a compound of general formula (I ) according to any one of claims 1 to 5, said method comprising the step of allowing an intermediate compound of general formula (VI I) :
Figure imgf000127_0002
in which X2, R4, R5, R7, and R8 are as defined for the compound of general formula (I ) in any one of claims 1 to 5,
to react with a compound of general formula (4- 1 ) :
Figure imgf000128_0001
4-1 in which R' , and R2 are as defined for the compound of general formula (I ) in any one of claims 1 to 5,
thereby giving a compound of general formula (I ) :
Figure imgf000128_0002
in which X1 represents 0, and , X2, R! , R2, R4, R5, R7, and R8 are as defined for the compound of general formula (I) in any one of claims 1 to 5.
1 1 . A compound of general formula (I ), or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, according to any one of claims 1 to 5, for use in the treatment or prophylaxis of a disease.
12. A pharmaceutical composition comprising a compound of general formula (I ), or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof , or a mixture of same, according to any one of claims 1 to 5, and a pharmaceutically acceptable diluent or carrier.
1 3. A pharmaceutical combination comprising :
one or more first active ingredients selected from a compound of general formula (I ) according to any of claims 1 to 5, and
one or more second active ingredients selected from chemotherapeutic anti-cancer agents.
14. Use of a compound of general formula (I ), or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof , or a mixture of same, according to any one of claims 1 to 5, for the prophylaxis or treatment of a disease.
1 5. Use of a compound of general formula (I ), or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof , or a mixture of same, according to any one of claims 1 to 5, for the preparation of a medicament for the prophylaxis or treatment of a disease.
16. Use according to claim 1 1 , 1 , or 1 5, wherein said disease is a disease of uncontrolled cell growth , proliferation and/or survival, an inappropriate cellular immune response, or an inappropriate cellular inflammatory response, particularly in which the disease of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune response, or inappropriate cellular inflammatory response is a haematological tumour, a solid tumour and/or metastases thereof, e.g. leukaemias and myelodysplastic syndrome, malignant lymphomas, head and neck tumours including brain tumours and brain metastases, tumours of the thorax including non-small cell and small cell lung tumours, gastrointestinal tumours, endocrine tumours, mammary and other gynaecological tumours, urological tumours including renal, bladder and prostate tumours, skin tumours, and sarcomas, and/or metastases thereof.
17. A compound or a salt thereof selected from :
Figure imgf000130_0001
in which Xi represents N , R\ and R2 are as defined for the compound of general formula (I ) in any one of claims 1 to 5;
Figure imgf000130_0002
in which Xi represents NR3, X2, R! , R2, R3, R4, R5, and R9 are as defined for the compound of general formula (I ) in any one of claims 1 to 5 and R13 represents R9 or H ;
Figure imgf000130_0003
in which Xi represents NR3, X2, R2, R3, R4, R5, R7 and R8 are as defined for the compound of general formula (I ) in any one of claims 1 to 5;
Figure imgf000131_0001
in which X1 represents N , and X2, R2, R4, R5, R7 and R8 are as defined for the compound of general formula (I ) in any one of claims 1 to 5; and
Figure imgf000131_0002
in which X2, R4, R5, R7, and R8 are as defined for the compound of general formula (I ) in any one of claims 1 to 5.
18. Use of a compound or a salt thereof as defined in claim 17, for the preparation of a compound of general formula (I )
Figure imgf000131_0003
(I) or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N- oxide, tautomer or stereoisomer,
in which X1 , X2, R1 , R2, R4, R5, R7, and R8 are as defined for the compound of general formula (I) in any one of claims 1 to 5.
PCT/EP2016/073040 2015-10-01 2016-09-28 Amido-substituted azole compounds WO2017055313A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP15188027.5 2015-10-01
EP15188027 2015-10-01

Publications (1)

Publication Number Publication Date
WO2017055313A1 true WO2017055313A1 (en) 2017-04-06

Family

ID=54252157

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2016/073040 WO2017055313A1 (en) 2015-10-01 2016-09-28 Amido-substituted azole compounds

Country Status (1)

Country Link
WO (1) WO2017055313A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9884063B2 (en) 2014-04-02 2018-02-06 Bayer Pharma Aktiengesellschaft Amido-substituted azole compounds
WO2018078005A1 (en) 2016-10-29 2018-05-03 Bayer Pharma Aktiengesellschaft Amido-substituted azaspiro derivatives as tankyrase inhibitors
WO2018078009A1 (en) 2016-10-29 2018-05-03 Bayer Pharma Aktiengesellschaft Amido-substituted cyclohexane derivatives
WO2018087126A1 (en) 2016-11-09 2018-05-17 Bayer Pharma Aktiengesellschaft Amido-substituted cyclohexane derivatives as inhibitors of tankyrase
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors

Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0023045A1 (en) * 1979-07-20 1981-01-28 Ajinomoto Co., Inc. Imidazolecarboxylic acid derivatives of penicillins and cephalosporins
US5011472A (en) 1988-09-06 1991-04-30 Brown University Research Foundation Implantable delivery system for biological factors
US5023252A (en) 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
WO2001000575A1 (en) 1999-06-24 2001-01-04 Nihon Nohyaku Co., Ltd. Heterocyclic dicarboxylic acid diamide derivatives, agricultural/horticultural insecticides and method of using the same
WO2008042282A2 (en) * 2006-09-28 2008-04-10 Exelixis, Inc. Imidazole-4, 5-dicarboxamide derivatives as jak-2 modulators
WO2008042283A2 (en) 2006-09-28 2008-04-10 Network Appliance, Inc. Write-in-place within a write-anywhere filesystem
WO2009059994A2 (en) 2007-11-05 2009-05-14 Novartis Ag Methods and compositions for measuring wnt activation and for treating wnt-related cancers
WO2012076898A1 (en) 2010-12-08 2012-06-14 Oslo University Hospital Hf Triazole derivatives as wnt signaling pathway inhibitors
WO2013010092A1 (en) 2011-07-13 2013-01-17 Novartis Ag 4-oxo-3,5,7,8-tetrahydro-4h-pyrano {4,3-d} pyrminidinyl compounds for use as tankyrase inhibitors
WO2013012723A1 (en) 2011-07-13 2013-01-24 Novartis Ag Novel 2-piperidin-1-yl-acetamide compounds for use as tankyrase inhibitors
WO2013093508A2 (en) 2011-12-22 2013-06-27 Oslo University Hospital Hf Wnt pathway inhibitors
WO2013134079A1 (en) 2012-03-05 2013-09-12 Amgen Inc. Oxazolidinone compounds and derivatives thereof
WO2013164061A1 (en) 2012-05-04 2013-11-07 dedeMERCK PATENT GMBH Pyrrolotriazinone derivatives
WO2013177349A2 (en) 2012-05-25 2013-11-28 Glaxosmithkline Llc Quinazolinediones as tankyrase inhibitors
WO2013182546A1 (en) 2012-06-07 2013-12-12 F. Hoffmann-La Roche Ag Pyrazolopyrimidone and pyrazolopyridone inhibitors of tankyrase
WO2013189905A1 (en) 2012-06-20 2013-12-27 F. Hoffmann-La Roche Ag Pyrrolopyrazone inhibitors of tankyrase
WO2013189865A1 (en) 2012-06-20 2013-12-27 F. Hoffmann-La Roche Ag N-aryltriazole compounds as lpar antagonists
WO2014023390A2 (en) 2012-08-08 2014-02-13 Merck Patent Gmbh (aza-)isoquinolinone derivatives
WO2015150449A2 (en) 2014-04-02 2015-10-08 Bayer Pharma Aktiengesellschaft Amido-substituted azole compounds

Patent Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0023045A1 (en) * 1979-07-20 1981-01-28 Ajinomoto Co., Inc. Imidazolecarboxylic acid derivatives of penicillins and cephalosporins
US5023252A (en) 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US5011472A (en) 1988-09-06 1991-04-30 Brown University Research Foundation Implantable delivery system for biological factors
WO2001000575A1 (en) 1999-06-24 2001-01-04 Nihon Nohyaku Co., Ltd. Heterocyclic dicarboxylic acid diamide derivatives, agricultural/horticultural insecticides and method of using the same
WO2008042282A2 (en) * 2006-09-28 2008-04-10 Exelixis, Inc. Imidazole-4, 5-dicarboxamide derivatives as jak-2 modulators
WO2008042283A2 (en) 2006-09-28 2008-04-10 Network Appliance, Inc. Write-in-place within a write-anywhere filesystem
WO2009059994A2 (en) 2007-11-05 2009-05-14 Novartis Ag Methods and compositions for measuring wnt activation and for treating wnt-related cancers
WO2012076898A1 (en) 2010-12-08 2012-06-14 Oslo University Hospital Hf Triazole derivatives as wnt signaling pathway inhibitors
WO2013010092A1 (en) 2011-07-13 2013-01-17 Novartis Ag 4-oxo-3,5,7,8-tetrahydro-4h-pyrano {4,3-d} pyrminidinyl compounds for use as tankyrase inhibitors
WO2013012723A1 (en) 2011-07-13 2013-01-24 Novartis Ag Novel 2-piperidin-1-yl-acetamide compounds for use as tankyrase inhibitors
WO2013093508A2 (en) 2011-12-22 2013-06-27 Oslo University Hospital Hf Wnt pathway inhibitors
WO2013134079A1 (en) 2012-03-05 2013-09-12 Amgen Inc. Oxazolidinone compounds and derivatives thereof
WO2013164061A1 (en) 2012-05-04 2013-11-07 dedeMERCK PATENT GMBH Pyrrolotriazinone derivatives
WO2013177349A2 (en) 2012-05-25 2013-11-28 Glaxosmithkline Llc Quinazolinediones as tankyrase inhibitors
WO2013182546A1 (en) 2012-06-07 2013-12-12 F. Hoffmann-La Roche Ag Pyrazolopyrimidone and pyrazolopyridone inhibitors of tankyrase
WO2013189905A1 (en) 2012-06-20 2013-12-27 F. Hoffmann-La Roche Ag Pyrrolopyrazone inhibitors of tankyrase
WO2013189865A1 (en) 2012-06-20 2013-12-27 F. Hoffmann-La Roche Ag N-aryltriazole compounds as lpar antagonists
WO2014023390A2 (en) 2012-08-08 2014-02-13 Merck Patent Gmbh (aza-)isoquinolinone derivatives
WO2015150449A2 (en) 2014-04-02 2015-10-08 Bayer Pharma Aktiengesellschaft Amido-substituted azole compounds

Non-Patent Citations (57)

* Cited by examiner, † Cited by third party
Title
ACS MED CHEM LETT, vol. 4, no. 12, 2013, pages 1173
ACS MED CHEM LETT, vol. 4, no. 12, 2013, pages 1218
ACS MED CHEM LETT, vol. 6, no. 3, 2014, pages 254
ACTA CRYSTALLOGR SECT F STRUCT BIOL CRYST COMMUN, vol. 68, 2012, pages 115
AIELLO ET AL., NEW ENGL. J. MED., vol. 331, 1994, pages 1480
ANGERS S; MOON RT.: "Proximal events in Wnt signal transduction", NAT REV MOL CELL BIOL., vol. 10, 2009, pages 468
BROWN, J.A.; MARALA, R.B., J. PHARMACOL. TOXICOL. METHODS, vol. 47, 2002, pages 137
CADIGAN KM; LIU YI.: "Wnt signaling: complexity at the surface.", J CELL SCI., vol. 119, 2006, pages 395, XP003020092, DOI: doi:10.1242/jcs.02826
CANCER RESEARCH, vol. 73, no. 10, 2013, pages 3132
CHEMMEDCHEM, vol. 8, no. 12, 2013, pages 1978
CURTIN JC; LORENZI MV.: "Drug Discovery Approaches to Target Wnt Signaling in Cancer Stem Cells", ONCOTARGET, vol. 1, 2010, pages 552
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 19 March 2003 (2003-03-19), XP002763804, retrieved from STN Database accession no. 499986-52-6 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 20 December 2007 (2007-12-20), XP002763805, retrieved from STN Database accession no. 958939-58-7 *
DONIGIAN JR; DE LANGE T.: "The role of the poly(ADP-ribose) polymerase tankyrase 1 in telomere length control by the TRF1 component of the shelterin complex.", J BIOL CHEM, vol. 282, 2007, pages 22662
DREGALLA RC; ZHOU J; IDATE RR; BATTAGLIA CL; LIBER HL,; BAILEY SM.: "Regulatory roles of tankyrase 1 at telomeres and in DNA repair: suppression of T-SCE and stabilization of DNA-PKcs", AGING, vol. 2, no. 10, 2010, pages 691
EAVES CJ; HUMPHRIES RK.: "Acute myeloid leukemia and the Wnt pathway", N ENGL J MED., vol. 362, 2010, pages 2326
GORDON MD; NUSSE R.: "Wnt signaling: multiple pathways, multiple receptors, and multiple transcription factors", J BIOL CHEM., vol. 281, 2006, pages 22429, XP003020091, DOI: doi:10.1074/jbc.R600015200
HE X; SEMENOV M; TAMAI K; ZENG X.: "LDL receptor-related proteins 5 and 6 in Wnt/beta-catenin signaling: arrows point the way", DEVELOPMENT, vol. 131, 2004, pages 1663, XP055298787, DOI: doi:10.1242/dev.01117
HUANG H; HE X.: "Wnt/beta-catenin signaling: new (and old) players and new insights.", CURR OPIN CELL BIOL., vol. 20, 2008, pages 119, XP022593569, DOI: doi:10.1016/j.ceb.2008.01.009
HUANG SMA; MISHINA YM; LIU S; CHEUNG A; STEGMEIER F ET AL.: "Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling", NATURE, vol. 461, 2009, pages 614, XP055062115, DOI: doi:10.1038/nature08356
J MED CHEM, vol. 55, no. 3, 2012, pages 1360
J MED CHEM, vol. 56, no. 11, 2013, pages 4320
J MED CHEM, vol. 56, no. 16, 2013, pages 6495
J MED CHEM, vol. 56, no. 17, 2013, pages 7049
J MED CHEM, vol. 56, no. 20, 2013, pages 7880
J MED CHEM, vol. 56, no. 24, 2013, pages 10003
J MED CHEM, vol. 56, no. 3, 2013, pages 1341
J MED CHEM, vol. 56, no. 7, 2013, pages 3012
KIMELMAN D; XU W.: "beta-catenin destruction complex: insights and questions from a structural perspective", ONCOGENE, vol. 25, 2006, pages 7482, XP002478898, DOI: doi:10.1038/sj.onc.1210055
KLAUS A; BIRCHMEIER W.: "Wnt signalling and its impact on development and cancer", NAT REV CANCER, vol. 8, 2008, pages 387 - 398, XP055123786, DOI: doi:10.1038/nrc2389
KLAUS A; BIRCHMEIERW.: "Wnt signalling and its impact on development and cancer", NAT REV CANCER, vol. 8, 2008, pages 387, XP055123786, DOI: doi:10.1038/nrc2389
L. N. VOSTROVA ET AL: "SYNTHESIS AND ANTIMICROBIAL ACTIVITY OF 5-NITROFURFURAL HETERYL HYDRAZONES", PHARMACEUTICAL CHEMISTRY JOURNAL, vol. 23, no. 5, 1 January 1989 (1989-01-01), pages 408 - 412, XP055315691 *
LEHTI L; CHI N-W; KRAUSS S.: "Tankyrases as drug targets", FEBS JOURNAL, vol. 280, 2013, pages 3576
LI Z,; YAMAUCHI Y; KAMAKURA M; MURAYAMA T; GOSHIMA F; KIMURA H; NISHIYAMA Y; HERPES SIMPLEX: "Virus Requires Poly(ADP-Ribose) Polymerase Activity for Efficient Replication and Induces Extracellular Signal-Related Kinase-Dependent Phosphorylation and ICPO-Dependent Nuclear Localization of Tankyrase 1", JOURNAL OF VIROLOGY, vol. 86, no. 1, 2012, pages 492
LIOTTA LA; KOHN EC.: "The microenvironment of the tumour-host interface", NATURE, vol. 411, 2001, pages 375, XP002362466, DOI: doi:10.1038/35077241
LOPEZ ET AL., INVEST. OPTHTHALMOL. VIS. SCI., vol. 37, 1996, pages 855
MEACHAM CE; MORRISON SJ.: "Tumour heterogeneity and cancer cell plasticity.", NATURE, vol. 501, 2013, pages 328
MOLENAAR ET AL., CELL, vol. 86, no. 3, 1996, pages 391 - 399
MORIN, SCIENCE, vol. 275, 1997, pages 1787 - 1790
NEMA, S. ET AL.: "Excipients and Their Use in Injectable Products", PDA JOURNAL OF PHARMACEUTICAL SCIENCE ET TECHNOLOGY, vol. 51, no. 4, 1997, pages 166 - 171
NUSSE R; FUERER C; CHING W; HARNISH K; LOGAN C; ZENG A; TEN BERGE D; KALANI Y.: "Wnt signaling and stem cell control", COLD SPRING HARB SYMP QUANT BIOL., vol. 73, 2008, pages 59
PEER ET AL., LAB. INVEST., vol. 72, 1995, pages 638
PERCHELLET ELISABETH M ET AL: "Imidazole-4,5-dicarboxamide Derivatives with Antiproliferative Activity against HL-60 Cells", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 48, no. 19, 1 January 2005 (2005-01-01), pages 5955 - 5965, XP002470716, ISSN: 0022-2623, DOI: 10.1021/JM050160R *
POLAKIS P.: "The many ways of Wnt in cancer", CURR OPIN GENET DEV., vol. 17, 2007, pages 45, XP005736994, DOI: doi:10.1016/j.gde.2006.12.007
POWELL, M.F. ET AL.: "Compendium of Excipients for Parenteral Formulations", PDA JOURNAL OF PHARMACEUTICAL SCIENCE ET TECHNOLOGY, vol. 52, no. 5, 1998, pages 238 - 311, XP009119027
PURE APPL CHEM, vol. 45, 1976, pages 11 - 30
RAO TP; KUHL M.: "An updated overview on Wnt signaling pathways: a prelude for more", CIRC RES., vol. 106, 2010, pages 1798
REYA T; CLEVERS H.: "Wnt signalling in stem cells and cancer", NATURE, vol. 434, 2005, pages 843, XP055229280, DOI: doi:10.1038/nature03319
RIJSEWIJK F; SCHUERMANN M; WAGENAAR E; PARREN P; WEIGEL D; NUSSE R.: "The Drosophila homolog of the mouse mammary oncogene int-1 is identical to the segment polarity gene wingless.", CELL, vol. 50, 1987, pages 649, XP023883197, DOI: doi:10.1016/0092-8674(87)90038-9
S. M. BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI, vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
SHAHI P; PARK D; POND AC; SEETHAMMAGARI M; CHIOU S-H; CHO K ET AL.: "Activation of Wnt signaling by chemically induced dimerization of LRP5 disrupts cellular homeostasis.", PLOS ONE, vol. 7, 2012, pages E30814
STRICKLEY, R.G: "Parenteral Formulations of Small Molecule Therapeutics Marketed in the United States (1999)-Part-1", PDA JOURNAL OF PHARMACEUTICAL SCIENCE ET TECHNOLOGY, vol. 53, no. 6, 1999, pages 324 - 349
T.W. GREENE; P.G.M. WUTS: "Protective Groups in Organic Synthesis", 1999, WILEY
VERMEULEN L; DE SOUSA EMF; VAN DER HEIJDEN M; CAMERON K; DE JONG JH; BOROVSKI T; TUYNMAN JB; TODARO M; MERZ C; RODERMOND H: "Wnt activity defines colon cancer stem cells and is regulated by the microenvironment", NAT CELL BIOL, vol. 12, 2010, pages 468
YASUDA N: "Synthesis of novel imidazole-4,5-dicarboxylic acid derivatives", JOURNAL OF HETEROCYCLIC CHEMISTRY, WILEY-BLACKWELL PUBLISHING, INC, US, vol. 22, 1 January 1985 (1985-01-01), pages 413 - 416, XP002275748, ISSN: 0022-152X, DOI: 10.1002/JHET.5570220239 *
YING Y. ET AL.: "Epigenetic disruption of the WNT/beta-catenin signaling pathway in human cancers", EPIGENETICS, vol. 4, 2009, pages 307
ZHANG Y; LIU S; MICKANIN C; FENG Y; CHARLAT 0 ET AL.: "RNF146 is a poly(ADP-ribose)-directed E3 ligase that regulates axin degradation and Wnt signalling", NATURE CELL BIOLOGY, vol. 13, 2011, pages 623

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9884063B2 (en) 2014-04-02 2018-02-06 Bayer Pharma Aktiengesellschaft Amido-substituted azole compounds
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385130B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US11535670B2 (en) 2016-05-11 2022-12-27 Huyabio International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
WO2018078005A1 (en) 2016-10-29 2018-05-03 Bayer Pharma Aktiengesellschaft Amido-substituted azaspiro derivatives as tankyrase inhibitors
WO2018078009A1 (en) 2016-10-29 2018-05-03 Bayer Pharma Aktiengesellschaft Amido-substituted cyclohexane derivatives
WO2018087126A1 (en) 2016-11-09 2018-05-17 Bayer Pharma Aktiengesellschaft Amido-substituted cyclohexane derivatives as inhibitors of tankyrase

Similar Documents

Publication Publication Date Title
EP3325451B1 (en) Fused imidazoles as midh1 inhibitors
EP3319945B1 (en) 2-aryl- and 2-arylalkyl-benzimidazoles as midh1 inhibitors
EP3303302B1 (en) N-menthylbenzimidazoles as midh1 inhibitors
AU2016257302A1 (en) Amido-substituted cyclohexane derivatives
EP3209646B1 (en) Benzimidazol-2-amines as midh1 inhibitors
CA2992364A1 (en) 5-hydroxyalkylbenzimidazoles as midh1 inhibitors
WO2017055313A1 (en) Amido-substituted azole compounds
EP3209660B1 (en) 1-cyclohexyl-2-phenylaminobenzimidazoles as midh1 inhibitors for the treatment of tumors
US9884063B2 (en) Amido-substituted azole compounds
WO2017055316A1 (en) Amido-substituted azole compounds
WO2016202758A1 (en) Substituted 2-(1h-pyrazol-1-yl)-1h-benzimidazole compounds
WO2016131794A1 (en) 3-carbamoylphenyl-4-carboxamide and isophtalamide derivatives as inhibitors of the wnt signalling pathway
WO2016202756A1 (en) Substituted 2-(1h-pyrazol-1-yl)-1h-benzimidazole compounds
WO2018087126A1 (en) Amido-substituted cyclohexane derivatives as inhibitors of tankyrase
WO2018078009A1 (en) Amido-substituted cyclohexane derivatives
WO2018078005A1 (en) Amido-substituted azaspiro derivatives as tankyrase inhibitors
WO2016202759A1 (en) Cytotoxic substituted 2-(1 h-pyrazol-1 -yl)-1,3-benzothiazole compounds for the treatment of cancer

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: 16770944

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: 16770944

Country of ref document: EP

Kind code of ref document: A1