WO2017157992A1 - Annulated pyrazoles as bub1 kinase inhibitors for treating proliferative disorders - Google Patents

Annulated pyrazoles as bub1 kinase inhibitors for treating proliferative disorders Download PDF

Info

Publication number
WO2017157992A1
WO2017157992A1 PCT/EP2017/056076 EP2017056076W WO2017157992A1 WO 2017157992 A1 WO2017157992 A1 WO 2017157992A1 EP 2017056076 W EP2017056076 W EP 2017056076W WO 2017157992 A1 WO2017157992 A1 WO 2017157992A1
Authority
WO
WIPO (PCT)
Prior art keywords
independently
alkyl
alkoxy
group selected
represent
Prior art date
Application number
PCT/EP2017/056076
Other languages
French (fr)
Inventor
Anne Mengel
Marion Hitchcock
Lars BÄRFACKER
Arwed Cleve
Hans Briem
Gerhard Siemeister
Amaury Ernesto FERNANDEZ-MONTALVAN
Jens SCHRÖDER
Ursula MÖNNING
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 WO2017157992A1 publication Critical patent/WO2017157992A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/052Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being six-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • the invention relates to substituted annulated pyrazole compounds, a process for their production and the use thereof.
  • the eukaryotic cell division cycle ensures the duplication of the genome and its distribution to the daughter cells by passing through a coordinated and regulated sequence of events.
  • the cell cycle is divided into four successive phases: 1 .
  • the G1 phase represents the time before the DNA replication, in which the cell grows and is sensitive to external stimuli.
  • the passage through the cell cycle is strictly regulated and controlled.
  • the enzymes that are necessary for the progression through the cycle must be activated at the correct time and are also turned off again as soon as the corresponding phase is passed.
  • Corresponding control points stop or delay the progression through the cell cycle if DNA damage is detected, or the DNA replication or the creation of the spindle device is not yet completed.
  • the mitotic checkpoint also known as spindle checkpoint or spindle assembly checkpoint
  • the mitotic checkpoint is active as long as unattached kinetochores are present and generates a wait-signal to give the dividing cell the time to ensure that each kinetochore is attached to a spindle pole, and to correct attachment errors.
  • the mitotic checkpoint prevents a mitotic cell from completing cell division with unattached or erroneously attached chromosomes [Suijkerbuijk SJ and Kops GJ, Biochem. Biophys. Acta 1786, 24, 2008; Musacchio A and Salmon ED, Nat. Rev. Mol. Cell. Biol. 8, 379, 2007].
  • the mitotic checkpoint is established by a complex network of a number of essential proteins, including members of the MAD (mitotic arrest deficient, MAD 1 -3) and Bub (Budding uninhibited by benzimidazole, Bub 1 -3) families, Mps1 kinase, cdc20, as well as other components [reviewed in Bolanos-Garcia VM and Blundell TL, Trends Biochem. Sci. 36, 141 , 2010], many of these being over-expressed in proliferating cells (e.g. cancer cells) and tissues [Yuan B et al., Clin. Cancer Res. 12, 405, 2006].
  • the major function of an unsatisfied mitotic checkpoint is to keep the anaphase-promoting complex/cyclosome (APC/C) in an inactive state.
  • APC/C anaphase-promoting complex/cyclosome
  • ubiquitin-ligase targets cyclin B and securin for proteolytic degradation leading to separation of the paired chromosomes and exit from mitosis.
  • Bub1 Inactive mutations of the Ser/Thr kinase Bub1 prevented the delay in progression through mitosis upon treatment of cells of the yeast S. cerevisiae with microtubule- destabilizing drugs, which led to the identification of Bub1 as a mitotic checkpoint protein [Roberts BT et al., Mol. Cell Biol., 14, 8282, 1994].
  • a number of recent publications provide evidence that Bub1 plays multiple roles during mitosis which, have been reviewed by Elowe [Elowe S, Mol. Cell. Biol. 31 , 3085, 201 1 ].
  • Bub1 is one of the first mitotic checkpoint proteins that binds to the kinetochores of duplicated chromosomes and probably acts as a scaffolding protein to constitute the mitotic checkpoint complex. Furthermore, via phosphorylation of histone H2A, Bub1 localizes the protein shugoshin to the centromeric region of the chromosomes to prevent premature segregation of the paired chromosomes [Kawashima et al. Science 327, 172, 2010]. In addition, together with a Thr-3 phosphorylated Histone H3 the shugoshin protein functions as a binding site for the chromosomal passenger complex which includes the proteins survivin, borealin, INCENP and Aurora B.
  • the chromosomal passenger complex is seen as a tension sensor within the mitotic checkpoint mechanism, which dissolves erroneously formed microtubule-kinetochor attachments such as syntelic (both sister kinetochors are attached to one spindle pole) or merotelic (one kinetochor is attached to two spindle poles) attachments [Watanabe Y, Cold Spring Harb. Symp. Quant. Biol. 75, 419, 2010].
  • Recent data suggest that the phosphorylation of histone H2A at Thr 121 by Bub1 kinase is sufficient to localize AuroraB kinase to fulfill the attachment error correction checkpoint [Ricke et al. J. Cell Biol. 199, 931 -949, 2012].
  • mitotic checkpoint abrogation through pharmacological inhibition of components of the mitotic checkpoint represents a new approach for the treatment of proliferative disorders, including solid tumours such as carcinomas, sarcomas, leukaemias and lymphoid malignancies or other disorders, associated with uncontrolled cellular proliferation.
  • the present invention relates to chemical compounds that inhibit Bub1 kinase.
  • Established anti-mitotic drugs such as vinca alkaloids, taxanes or epothilones activate the mitotic checkpoint, inducing a mitotic arrest either by stabilising or destabilising microtubule dynamics. This arrest prevents separation of the duplicated chromosomes to form the two daughter cells. Prolonged arrest in mitosis forces a cell either into mitotic exit without cytokinesis (mitotic slippage or adaption) or into mitotic catastrophe leading to cell death [Rieder CL and Maiato H, Dev. Cell 7, 637, 2004].
  • Bub1 inhibitors should be of therapeutic value for the treatment of proliferative disorders associated with enhanced uncontrolled proliferative cellular processes such as, for example, cancer, inflammation, arthritis, viral diseases, cardiovascular diseases, or fungal diseases in a warm-blooded animal such as man.
  • WO 2013/050438, WO 2013/092512, WO 2013/167698 disclose substituted benzylindazoles, substituted benzylpyrazoles and substituted benzylcycloalkyl- pyrazoles, respectively, which are Bub1 kinase inhibitors.
  • WO 2014/147203, WO 2014/147204, WO2014202590, WO2014202588, WO2014202584, WO2014202583, and WO2015/063003, disclose substituted indazoles, substituted pyrazoles, and substituted cycloalkylpyrazoles, which are Bub1 kinase inhibitors.
  • Inhibitors of Bub1 represent valuable compounds that should complement therapeutic options either as single agents or in combination with other drugs.
  • the invention covers compounds of general formula (I),
  • V, W, Y and Z independently of each other, represent CH or CR 5 ,
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , or,
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 , or,
  • V and Y represent N, and W and Z, independently of each other, represent CH or CR 5 ,
  • R 1 and R 2 represent, independently of each other, a hydrogen atom or a halogen atom
  • R 3 represents a hydrogen atom or a group selected from:
  • Ci-C 6 -alkyl Ci-C 6 -haloalkyl, Ci-C 6 -hydroxyalkyl
  • hydroxyalkyl groups are optionally substituted with one, two or three halogen atoms selected from:
  • fluorine and chlorine represents a hydrogen atom, or a group selected from:
  • Ci-Ce-alkyl C3-C 6 -cycloalkyl, Ci-C 6 -haloalkyl, C 2 -C 6 -hydroxyalkyl and
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • Ci-C3-alkyl Ci-C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy,
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group
  • Ci-C 3 -alkyl Ci-C 3 -haloalkyl, C 2 -C 3 -hydroxyalkyl, C3-C 4 -cycloalkyl,
  • R10 represents a hydrogen atom or a group selected from:
  • Ci-C 3 -alkyl Ci-C 3 -haloalkyl, C 2 -C 3 -hydroxyalkyl, C3-C 4 -cycloalkyl,
  • R 1 1 represents, independently of each other, a hydrogen atom or a halogen atom or a group selected from:
  • a second aspect of the invention are compounds of formula (I) as defined herein, wherein
  • V, W, Y and Z independently of each other, represent CH or CR 5 ,
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , or,
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 ,
  • R 1 and R 2 represent, independently of each other, a hydrogen atom or a halogen atom
  • R 3 represents a hydrogen atom or a group selected from:
  • R 4 represents a hydrogen atom, or a group selected from:
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • Ci -C 3 -alkyl, C3-C 4 -cycloalkyl, Ci -C 3 -haloalkyl, Ci -C 3 -alkoxy, C1 -C3- haloalkoxy and -C( 0)N(R 9 ) 2 ,
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from:
  • R 11 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group, or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
  • a third aspect of the invention are compounds of formula (I) as defined herein, wherein
  • V, W, Y and Z independently of each other, represent CH or CR 5 ,
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , or,
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 , R 1 and R 2 represent, independently of each other, a hydrogen atom or a halogen atom, R 3 represents a hydrogen atom or a group selected from:
  • Ci-C3-alkoxy (C2-C3-hydroxyalkyl)-0- and (Ci-C3-alkoxy)-(C2-C3-alkoxy)-,
  • R 4 represents a hydrogen atom, or a group selected from:
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from:
  • Ci-C 3 -alkyl and C3-C 4 -cycloalkyl are Ci-C 3 -alkyl and C3-C 4 -cycloalkyl
  • R 11 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group, or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
  • a fourth aspect of the invention are compounds of formula (I) as defined herein, wherein V, W, Y and Z, independently of each other, represent CH or CR 5 ,
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , or,
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 ,
  • R 1 and R 2 represent, independently of each other, a halogen atom
  • R 3 represents a group selected from:
  • R 4 represents a
  • R 5 represents, independently of each other, a group selected from:
  • Ci-C3-alkyl and Ci-C3-alkoxy are Ci-C3-alkyl and Ci-C3-alkoxy
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group
  • a fifth aspect of the invention are compounds of formula (I) as defined herein, wherein V, W, Y and Z, independently of each other, represent CH or CR 5 ,
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , or,
  • W represents N, and V, Y and Z, independently of each other, represent CH,
  • R 1 and R 2 represent a fluorine atom
  • R 3 represents a group selected from:
  • R 4 represents a methyl group
  • R 5 represents, independently of each other, a group selected from:
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group
  • a further aspect of the invention are compounds of formula (I), wherein
  • V, W, Y and Z independently of each other, represent CH or CR 5 ,
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , or,
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 , or,
  • V and Y represent N, and W and Z, independently of each other, represent CH or CR 5 and
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • Ci-C3-alkyl Ci-C4-cycloalkyl
  • Ci-C3-haloalkyl Ci-C3-alkoxy
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from:
  • Ci-C 3 -alkyl Ci-C 3 -haloalkyl, C 2 -C3-hydroxyalkyl, C3-C 4 -cycloalkyl,
  • R 10 represents a hydrogen atom or a group selected from:
  • Ci-C 3 -alkyl Ci-C 3 -haloalkyl, C 2 -C 3 -hydroxyalkyl, C 3 -C 4 -cycloalkyl,
  • V, W, Y and Z independently of each other, represent CH or CR 5 ,
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , or,
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 , and
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from:
  • Ci-C 3 -alkyl C 2 -C 3 -hydroxyalkyl, C 3 -C -cycloalkyl and (Ci-C 3 -alkoxy)-(C 2 -C 3 - alkyl)-.
  • V, W, Y and Z independently of each other, represent CH or CR 5 ,
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , or,
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 , and R 5 represents, independently of each other, a halogen atom or a group selected from:
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from:
  • Ci-C3-alkyl and C3-C4-cycloalkyl are Ci-C3-alkyl and C3-C4-cycloalkyl.
  • V, W, Y and Z independently of each other, represent CH or CR 5 ,
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , or,
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 , and
  • R 5 represents, independently of each other, a group selected from:
  • Ci-C3-alkyl and Ci-C3-alkoxy are Ci-C3-alkyl and Ci-C3-alkoxy.
  • V, W, Y and Z independently of each other, represent CH or CR 5 ,
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , or,
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 , and
  • R 5 represents, independently of each other, a group selected from:
  • a further aspect of the invention are compounds of formula (I), wherein
  • V, W, Y and Z independently of each other, represent CH or CR 5 .
  • V, W, Y and Z represents CR 5 , and the others represent CH.
  • V represents N, and one of W, Y and Z, represents CR 5 , and the others represent CH.
  • a further aspect of the invention are compounds of formula (I), wherein
  • V, W, Y and Z independently of each other, represent CH or CR 5 ,
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • Ci-C 3 -alkyl C 3 -C 4 -cycloalkyl
  • Ci-C 3 -haloalkyl Ci-C 3 -alkoxy
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from :
  • Ci-C 3 -alkyl Ci-C 3 -haloalkyl, C 2 -C 3 -hydroxyalkyl, C 3 -C 4 -cycloalkyl,
  • R 10 represents a hydrogen atom or a group selected from :
  • V, W, Y and Z independently of each other, represent CH or CR 5 ,
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from :
  • Ci-C 3 -alkyl C 2 -C 3 -hydroxyalkyl, C 3 -C -cycloalkyl and (Ci-C3-alkoxy)-(C 2 -C 3 - alkyl)-.
  • Yet another aspect of the invention are compounds of formula (I), in which V, W, Y and Z, independently of each other, represent CH or CR 5 ,
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from:
  • Ci-C3-alkyl and C3-C4-cycloalkyl are Ci-C3-alkyl and C3-C4-cycloalkyl.
  • V, W, Y and Z independently of each other, represent CH or CR 5 ,
  • R 5 represents, independently of each other, a group selected from:
  • Ci-C 3 -alkyl and Ci-C 3 -alkoxy are examples of compounds that are identical to those that are used in the trade.
  • V, W, Y and Z independently of each other, represent CH or CR 5 ,
  • R 5 represents a methoxy group.
  • a further aspect of the invention are compounds of formula (I), wherein
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 .
  • a further aspect of the invention are compounds of formula (I), wherein
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , and
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • Ci-C 3 -alkyl C 3 -C 4 -cycloalkyl
  • Ci-C 3 -haloalkyl Ci-C 3 -alkoxy
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from:
  • Ci-C3-alkyl Ci-C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
  • R 10 represents a hydrogen atom or a group selected from:
  • Ci-C3-alkyl Ci-C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , and
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • Ci -C 3 -alkyl, C3-C 4 -cycloalkyl, Ci-C 3 -haloalkyl, Ci-C 3 -alkoxy, C1 -C3- haloalkoxy and -C( 0)N(R 9 ) 2 ,
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from:
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , and
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from:
  • Ci-C3-alkyl and C3-C4-cycloalkyl are Ci-C3-alkyl and C3-C4-cycloalkyl.
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , R 5 represents, independently of each other, a group selected from:
  • Ci-C 3 -alkyl and Ci-C 3 -alkoxy are examples of compounds that are identical to those that are used in the trade.
  • V represents N, and W, Y and Z, independently of each other, represent CH or CR 5 , and
  • R 5 represents a methyl group.
  • a further aspect of the invention are compounds of formula (I), wherein
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 .
  • a further aspect of the invention are compounds of formula (I), wherein
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 , and
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • Ci-C 3 -alkyl C3-C 4 -cycloalkyl
  • Ci-C 3 -haloalkyl Ci-C 3 -alkoxy
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from:
  • Ci-C 3 -alkyl Ci-C 3 -haloalkyl, C 2 -C 3 -hydroxyalkyl, C 3 -C 4 -cycloalkyl,
  • R 10 represents a hydrogen atom or a group selected from:
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 , and
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from:
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 , and
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from:
  • Ci-C 3 -alkyl and C 3 -C 4 -cycloalkyl are Ci-C 3 -alkyl and C 3 -C 4 -cycloalkyl.
  • W represents N, and V, Y and Z, independently of each other, represent CH or CR 5 , and
  • R 5 represents, independently of each other, a group selected from:
  • Ci -C 3 -alkyl and Ci-C 3 -alkoxy are examples of the same inorganic compounds.
  • W represents N
  • V, Y and Z represent CH.
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • Ci-C3-alkyl Ci-C4-cycloalkyl
  • Ci-C3-haloalkyl Ci-C3-alkoxy
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • Ci-C 3 -alkyl C3-C 4 -cycloalkyl
  • R 5 represents, independently of each other, a halogen atom or a group selected from:
  • R 5 represents, independently of each other, a group selected from :
  • Ci-C 3 -alkyl and Ci-C 3 -alkoxy are examples of compounds that are identical to those that are used in the trade.
  • R 5 represents, independently of each other, a group selected from :
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from :
  • Ci-C 3 -alkyl Ci-C 3 -haloalkyl, C 2 -C 3 -hydroxyalkyl, C 3 -C 4 -cycloalkyl,
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from :
  • R 9 represents, independently of each other, a hydrogen atom or a group selected from :
  • Ci-C 3 -alkyl and C 3 -C -cycloalkyl are compounds of formula (I), in which
  • R 9 represents, independently of each other, a hydrogen atom or a methyl group.
  • R 10 represents a hydrogen atom or a group selected from:
  • Ci -C3-alkyl Ci -C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
  • R 1 and R 2 represent, independently of each other, a halogen atom, preferably a fluorine atom.
  • R 1 and R 2 represent, independently of each other, a hydrogen atom or a halogen atom
  • R 3 represents a hydrogen atom or a group selected from:
  • Ci -Ce-alkyl Ci -Ce-haloalkyl, Ci -Ce-hydroxyalkyl
  • hydroxyalkyl groups are optionally substituted with one, two or three halogen atoms selected from:
  • R 1 and R 2 represent, independently of each other, a hydrogen atom or a halogen atom
  • R 3 represents a hydrogen atom or a group selected from:
  • R 1 and R 2 represent, independently of each other, a hydrogen atom or a halogen atom
  • R 3 represents a hydrogen atom or a group selected from:
  • R 1 and R 2 represent, independently of each other, a halogen atom
  • R 3 represents a group selected from:
  • R 1 and R 2 represent a fluorine atom
  • R 3 represents a group selected from:
  • R 4 represents a hydrogen atom, or a group selected from:
  • Ci-Ce-alkyl C 3 -C 6 -cycloalkyl, Ci-C 6 -haloalkyl, C 2 -C 6 -hydroxyalkyl and
  • R 4 represents a hydrogen atom, or a group selected from:
  • Ci-C 3 -alkyl and C 2 -C 3 -hydroxyalkyl are Ci-C 3 -alkyl and C 2 -C 3 -hydroxyalkyl.
  • Yet another aspect of the invention are compounds of formula (I), in which represents a Ci-C 3 -alkyl group.
  • R 4 represents a methyl group.
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group selected from:
  • R 11 represents, independently of each other, a hydrogen atom or a halogen atom or a group selected from:
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group
  • R 1 1 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group.
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group
  • R 1 1 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group.
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group
  • R 1 1 represents, independently of each other, a hydrogen atom or a halogen atom or a group selected from :
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group
  • R 1 1 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group.
  • R 7 and R 8 are linked to one another in such a way that they jointly form a group
  • R 11 represents, independently of each other, a hydrogen atom or a halogen atom or a group selected from:
  • R 11 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group.
  • R 11 represents a hydrogen atom.
  • One aspect of the invention are compounds of formula (I) as described in the examples, as characterized by their names in the title, as claimed in claim 6, and their structures as well as the subcombinations of all residues specifically disclosed in the compounds of the examples.
  • Another aspect of the present invention are the intermediates as used for their synthesis.
  • a further aspect of the invention are compounds of formula (I), which are present as their salts.
  • the salt is a pharmaceutically acceptable salt. 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.
  • 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 embodiment of the invention are compounds according to the claims as disclosed in the Claims section wherein the definitions are limited according to the preferred or more preferred definitions as disclosed below or specifically disclosed residues of the exemplified compounds and subcombinations thereof.
  • the present invention relates to intermediate compounds which are useful in the preparation of compounds of the present invention of general formula (I), particularly in the method described herein.
  • the present invention relates to a compound of general formula (II) :
  • R 1 , R 2 , R 3 , R 4 , R 7 and R 8 are as defined herein, for the preparation of a compound of general formula (I) as defined supra.
  • the present invention relates to the use of an intermediate compound of general formula (III) :
  • V, W, Y and Z are as defined herein, and X 2 represents F, CI, Br, I, boronic acid or a boronic acid ester, such as for example 4,4,5,5-tetramethyl-2-phenyl-1 ,3,2- dioxaborolane (boronic acid pinacole ester), for the preparation of a compound of general formula (I) as defined supra.
  • X 2 represents F, CI, Br, I, boronic acid or a boronic acid ester, such as for example 4,4,5,5-tetramethyl-2-phenyl-1 ,3,2- dioxaborolane (boronic acid pinacole ester), for the preparation of a compound of general formula (I) as defined supra.
  • the present invention relates to a method of preparing a compound of general formula (I) as defined supra, said method comprising the step of allowing an intermediate compound of general formula (II) :
  • V, W, Y and Z are as defined herein, and X 2 represents F, CI, Br, I, boronic acid or a boronic acid ester, such as for example 4,4,5,5-tetramethyl-2-phenyl-1 ,3,2- dioxaborolane (boronic acid pinacole ester),
  • the present invention relates to a method of preparing a compound of general formula (l-b) as defined herein, said method comprising the step of allowing a compound of general formula (l-a) :
  • R 1 , R 2 , R 4 , R 7 , R 8 , V, W, Y and Z are as defined herein, and R 12 represents a methyl group to react with a reagent such as for example boron tribromide,
  • R 1 , R 2 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 1 1 , V, W, Y and/or Z occur more than one time for any compound of formula (I) each definition of R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 1 1 , V, W, Y and/or Z is independent.
  • an oxo substituent represents an oxygen atom, which is bound to a carbon atom or to a sulfur atom via a double bond.
  • a composite substituent be composed of more than one parts, e.g. (C1 -C3- alkoxy)-(Ci-C 6 -alkyl), (Cs-Ce-cycloalkylHCi-Cs-alkyl)- or (Cs-Ce-cycloalkylHCi-Cs- alkoxy)-, it is possible for the position of a given part to be at any suitable position of said composite substituent, i.e. the Cs-Ce-cycloalkyl part can be attached to any carbon atom of the Ci-C 3 -alkyl part of said (C3-C 6 -cycloalkyl)-(Ci -C3-alkyl)- group.
  • a hyphen at the beginning or at the end of such a composite substituent indicates the point of attachment of said composite substituent to the rest of the molecule.
  • a ring comprising carbon atoms and optionally one or more heteroatoms, such as nitrogen, oxygen or sulfur atoms for example, be substituted with a substituent, it is possible for said substituent to be bound at any suitable position of said ring, be it bound to a suitable carbon atom and/or to a suitable heteroatom.
  • halogen atom is to be understood as meaning a fluorine, chlorine, bromine or iodine atom.
  • Ci -C 6 -alkyl is to be understood as meaning a linear or branched, saturated, monovalent hydrocarbon group having 1 , 2, 3, 4, 5 or 6 carbon atoms, e.g.
  • said group has 1 , 2, 3, 4 or 5 carbon atoms ("Ci-C 5 -alkyl”), e.g. a methyl, ethyl, propyl, butyl, iso-propyl, iso-butyl, sec-butyl, tert-butyl, iso-pentyl, 2-methylbutyl, 1 -methylbutyl, 1 -ethylpropyl, 1 ,2-dimethylpropyl, neo-pentyl, 1 ,1 -dimethylpropyl group, more particularly 1 , 2 or 3 carbon atoms (“Ci-C3-alkyl”), e.g. a methyl, ethyl, n-propyl- or iso-propyl group.
  • Ci-C 5 -alkyl e.g. a methyl, ethyl, propyl, butyl, iso-propyl, iso-butyl,
  • Ci-C 6 -haloalkyl is to be understood as meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "Ci-C 6 -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.
  • Ci-Ce-haloalkyl group is, for example, -CF 3 , - CHF 2 , -CH 2 F, -CF2CF3, -CH2CH2F, -CH2CHF2, -CH2CF3, -CH2CH2CF3, or -CH(CH 2 F) 2 .
  • said group has 1 , 2, 3 or 4 carbon atoms ("Ci-C 4 -haloalkyl"), more particularly 1 , 2 or 3 carbon atoms (“Ci-C3-haloalkyl”), even more particularly 1 or 2 carbon atoms ("Ci-C2-haloalkyl").
  • Ci -C 6 -alkoxy is to be understood as meaning a linear or branched, saturated, monovalent, hydrocarbon group of formula -O-alkyl, in which the term “alkyl” is defined supra, e.g. a methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, iso-butoxy, tert-butoxy, sec-butoxy, pentoxy, iso-pentoxy, or n-hexoxy group, or an isomer thereof.
  • said group has 1 , 2, 3 or 4 carbon atoms ("Ci-C4-alkoxy”), more particularly 1 , 2 or 3 carbon atoms ("Ci-C 3 -alkoxy").
  • Ci-Ce-haloalkoxy is to be understood as meaning a linear or branched, saturated, monovalent Ci -C 6 -alkoxy group, as defined supra, in which one or more of the hydrogen atoms is replaced, in identically or differently, by a halogen atom.
  • said halogen atom is F.
  • Said Ci-C 6 -haloalkoxy group is, for example, - OCF3, -OCHF2, -OCH2F, -OCF2CF3, or -OCH2CF3.
  • said group has 1 , 2, 3 or 4 carbon atoms ("Ci-C 4 -haloalkoxy"), more particularly 1 , 2 or 3 carbon atoms ("C1-C3- haloalkoxy").
  • Ci-Ce-hydroxyalkyI is to be understood as preferably meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "Ci-C 6 -alkyl” 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, 3- hydroxy-2-methyl-propyl, 2-hydroxy-2-methyl-propyl, 1 -hydroxy-2-methyl-propyl group.
  • said group has 1 , 2, 3 or 4 carbon atoms ("Ci-C 4 -hydroxyalkyl"), more particularly 1 , 2 or 3 carbon atoms ("Ci-C3-hydroxyalkyl").
  • Cs-Ce-cycloalkyl is to be understood as meaning a saturated, monovalent, monocyclic hydrocarbon ring which contains 3, 4, 5 or 6 carbon atoms ("C 3 -C 6 - cycloalkyl").
  • Said C3-C 6 -cycloalkyl group is for example, a monocyclic hydrocarbon ring, e.g. a cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl ring. Particularly, said group has 3 or 4 carbon atoms (“C3-C4-cycloalkyl").
  • Cs-Ce-cycloalkoxy means a saturated, monovalent, monocyclic group of formula (C3-C 6 -cycloalkyl)-0-, which contains 3, 4, 5 or 6 carbon atoms, in which the term "Cs-Ce-cycloalkyl” is defined supra, e.g. a cyclopropyloxy, cyclobutyloxy, cyclopentyloxy or cyclohexyloxy group.
  • Ci-C 6 as used in the present text, e.g. in the context of the definition of "Ci-Ce-alkyl", “Ci-C 6 -haloalkyl", “Ci-Ce-hydroxyalkyI”, “Ci-C 6 -alkoxy” or “Ci-C 6 - haloalkoxy” means a group having a finite number of carbon atoms of 1 to 6, i.e. 1 , 2, 3, 4, 5 or 6 carbon atoms.
  • C3-C 6 as used in the present text, e.g. in the context of the definition of "C3-C 6 -cycloalkyl", means a cycloalkyl group having a finite number of carbon atoms of 3 to 6, i.e. 3, 4, 5 or 6 carbon atoms.
  • d-Ce encompasses Ci , C2, C3, C4, C5, Ce, C1 -C6, C1 -C5, C1 -C4, C1 -C3, C1 -C2, C2-C6, C2-C5, C2-C4, C2-C3, C3-C6, C3-C5, C3-C4, C4-C6, C4-C5, and C5-C6;
  • C1 -C3 encompasses Ci , C 2 , C 3 , C1 -C3, C1 -C2 and C2-C3;
  • C2-C6 encompasses C2, C3, C4, C5, Ce, C2-C6, C2-C5, C2-C4, C2-C3, C3-C6, C3-C5, C3-C4, C4-C6, C4-C5, and C5-C6;
  • C 3 -C 6 encompasses C 3 , C 4 , C 5 , C 6 , C 3 -C 6 , C3-C5, C 3 -C 4 , C 4 -C 6 , C 4 -C 5 and C 5 -C 6 ;
  • C4-C6 encompasses C 4 , C5, Ce, C4-C6, C4-C5 and Cs-Ce;
  • C 3 -C 6 encompasses C 3 , C 4 , C 5 , C 6 , C 3 -C 6 , C3-C5, C 3 -C 4 , C 4 -C 6 , C 4 -C 5 , and C 5 -C 6 .
  • the term "leaving group” means an atom or a group of atoms that is displaced in a chemical reaction as stable species taking with it the bonding electrons.
  • a leaving group is selected from the group comprising: halide, in particular fluoride, chloride, bromide or iodide, (methylsulfonyl)oxy, [(trifluoromethyl)sulfonyl]oxy, [(nonafluorobutyl)sulfonyl]oxy, (phenylsulfonyl)oxy, [(4-methylphenyl)sulfonyl]oxy, [(4-bromophenyl)sulfonyl]oxy, [(4-nitrophenyl)sulfonyl]- oxy, [(2-nitrophenyl)sulfonyl]oxy, [(4-isopropylphenyl)sulfonyl]oxy, [(2,4,6-triisopropy
  • 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 "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 therefore includes one or more isotopic variant(s) of the compounds of general formula (I), particularly deuterium-containing compounds of general formula (I).
  • isotopic variant of a compound or a reagent is defined as a compound exhibiting an unnatural proportion of one or more of the isotopes that constitute such a compound.
  • Isotopic variant of the compound of general formula (I) is defined as a compound of general formula (I) exhibiting an unnatural proportion of one or more of the isotopes that constitute such a compound.
  • unnatural proportion means a proportion of such isotope which is higher than its natural abundance.
  • the natural abundances of isotopes to be applied in this context are described in "Isotopic Compositions of the Elements 1997", Pure Appl. Chem., 70(1 ), 217-235, 1998.
  • isotopes examples include stable and radioactive isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine and iodine, such as 2 H (deuterium), 3 H (tritium), 11 C, 13 C, 14 C, 15 N, 17 0, 18 0, 32 P, 33 P, 33 S, 34 S, 35 S, 36 S, 18 F, 36 CI, 82 Br, 123 l, 124 l, 125 l, 129 l and 131 1, respectively.
  • isotopes include stable and radioactive isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine and iodine, such as 2 H (deuterium), 3 H (tritium), 11 C, 13 C, 14 C, 15 N, 17 0, 18 0, 32 P, 33 P, 33 S, 34 S, 35 S, 36 S, 18 F, 36 CI, 82 Br, 123 l, 124 l, 125 l
  • the isotopic variant(s) of the compounds of general formula (I) preferably contain deuterium ("deuterium-containing compounds of general formula (I)").
  • Isotopic variants of the compounds of general formula (I) in which one or more radioactive isotopes, such as 3 H or 14 C, are incorporated are useful e.g. in drug and/or substrate tissue distribution studies. These isotopes are particularly preferred for the ease of their incorporation and detectability.
  • Positron emitting isotopes such as 18 F or 11 C may be incorporated into a compound of general formula (I).
  • These isotopic variants of the compounds of general formula (I) are useful for in vivo imaging applications.
  • Deuterium-containing and 13 C- containing compounds of general formula (I) can be used in mass spectrometry analyses in the context of preclinical or clinical studies.
  • Isotopic variants of the compounds of general formula (I) can generally be prepared by methods known to a person skilled in the art, such as those described in the schemes and/or examples herein, by substituting a reagent for an isotopic variant of said reagent, preferably for a deuterium-containing reagent.
  • a reagent for an isotopic variant of said reagent preferably for a deuterium-containing reagent.
  • deuterium from D 2 0 can be incorporated either directly into the compounds or into reagents that are useful for synthesizing such compounds.
  • Deuterium gas is also a useful reagent for incorporating deuterium into molecules.
  • Catalytic deuteration of olefinic bonds and acetylenic bonds is a rapid route for incorporation of deuterium.
  • Metal catalysts i.e.
  • deuterated reagents and synthetic building blocks are commercially available from companies such as for example C/D/N Isotopes, Quebec, Canada; Cambridge Isotope Laboratories Inc., Andover, MA, USA; and CombiPhos Catalysts, Inc., Princeton, NJ, USA.
  • deuterium-containing compound of general formula (I) is defined as a compound of general formula (I), in which one or more hydrogen atom(s) is/are replaced by one or more deuterium atom(s) and in which the abundance of deuterium at each deuterated position of the compound of general formula (I) is higher than the natural abundance of deuterium, which is about 0.015%.
  • the abundance of deuterium at each deuterated position of the compound of general formula (I) is higher than 10%, 20%, 30%, 40%, 50%, 60%, 70% or 80%, preferably higher than 90%, 95%, 96% or 97%, even more preferably higher than 98% or 99% at said position(s). It is understood that the abundance of deuterium at each deuterated position is independent of the abundance of deuterium at other deuterated position(s).
  • the selective incorporation of one or more deuterium atom(s) into a compound of general formula (I) may alter the physicochemical properties (such as for example acidity [C. L. Perrin, et al., J. Am. Chem. Soc., 2007, 129, 4490], basicity [C. L. Perrin et al., J. Am. Chem. Soc, 2005, 127, 9641 ], lipophilicity [B. Testa et al., Int. J. Pharm., 1984, 19(3), 271 ]) and/or the metabolic profile of the molecule and may result in changes in the ratio of parent compound to metabolites or in the amounts of metabolites formed.
  • physicochemical properties such as for example acidity [C. L. Perrin, et al., J. Am. Chem. Soc., 2007, 129, 4490]
  • basicity C. L. Perrin et al., J. Am. Chem. Soc,
  • a compound of general formula (I) may have multiple potential sites of attack for metabolism.
  • deuterium-containing compounds of general formula (I) having a certain pattern of one or more deuterium-hydrogen exchange(s) can be selected.
  • the deuterium atom(s) of deuterium-containing compound(s) of general formula (I) is/are attached to a carbon atom and/or is/are located at those positions of the compound of general formula (I), which are sites of attack for metabolizing enzymes such as e.g. cytochrome P450.
  • the present invention concerns a deuterium-containing compound of general formula (I) having 1 , 2, 3 or 4 deuterium atoms, particularly with 1 , 2 or 3 deuterium atoms.
  • 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 the present invention optionally contain one or more asymmetric centres, depending upon the location and nature of the various substituents desired. It is possible that one or more asymmetric carbon atoms are present in the (R) or (S) configuration, which can result in racemic mixtures in the case of a single asymmetric centre, and in diastereomeric mixtures in the case of multiple asymmetric centres. In certain instances, it is possible that asymmetry 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.
  • 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, diacetyltartaric, 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.
  • 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.
  • S. M. Berge, et 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, bisulfuric, 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, per
  • 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.
  • the salts include water-insoluble and, particularly, water-soluble salts.
  • bioprecursors or pro-drugs are covered by the invention.
  • Said biological system is e.g. a mammalian organism, particularly a human subject.
  • the bioprecursor is, for example, converted into the compound of formula (I) or a salt thereof by metabolic processes.
  • 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, Ci-C 6 alkoxymethyl esters, e.g. methoxymethyl, Ci -C 6 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.
  • pharmacokinetic profile means one single parameter or a combination thereof including permeability, bioavailability, exposure, and pharmacodynamic parameters such as duration, or magnitude of pharmacological effect, as measured in a suitable experiment.
  • Compounds with improved pharmacokinetic profiles can, for example, be used in lower doses to achieve the same effect, may achieve a longer duration of action, or a may achieve a combination of both effects.
  • 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. Any such combination of a compound of formula (I) of the present invention with an anticancer agent as defined below is an embodiment of the invention.
  • (chemotherapeutic) anti-cancer agents includes but is not limited to :
  • said compounds of the present invention have surprisingly been found to effectively inhibit Bub1 kinase 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 is mediated by Bub1 kinase, 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 compounds according to the invention can be prepared according to the following schemes 1 through 9.
  • Scheme 1 Route for the preparation of compounds of general formula (I), wherein R 1 , R 2 , R 3 , R 4 , R 7 , R 8 , V, W, Y and Z have the meaning as given for general formula (I), supra,
  • X 1 represents F, CI, Br, I or a sulfonate, e.g. trifluormethylsulfonate or p- toluolsulfonate.
  • X 2 represents F, CI, Br, I, boronic acid or a boronic acid ester, such as for example 4, 4, 5, 5-tetramethyl-2-phenyl-1 ,3,2-dioxaborolane (boronic acid pinacole ester).
  • interconversion of any of the substituents R 1 , R 2 R 3 , R 4 , R 7 , R 8 , V, W, Y or Z 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, 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.
  • a suitably substituted 1 H-pyrazole-3-carboxylic acid of the general formula (1 -1 ) can be reacted with methanol or ethanol in the presence of catalytic amounts of a Broensted acid, such as, for example, hydrochloric acid or sulphuric acid, at temperatures ranging from 0 ⁇ to boiling point of the respective alcohol , preferably the reaction is carried out at 85 ⁇ , to furnish alkyl 1 H-indazole-3-carboxylat e intermediates of general formula (1 -2).
  • a Broensted acid such as, for example, hydrochloric acid or sulphuric acid
  • Intermediates of general formula (1 -5) can be converted to intermediates of general formula (1 -7) by reaction with a suitably substituted 3,3-bis- (dimethylamino)propanenitrile of the general formula (1 -6), such as, for example 3,3- bis(dimethylamino)-2-methoxypropanenitrile, in the presence of a suitable base, such as, for example piperidine, in a suitable solvent system, such as, for example, 3- methylbutan-1 -ol, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 100 ⁇ .
  • a suitably substituted 3,3-bis- (dimethylamino)propanenitrile of the general formula (1 -6), such as, for example 3,3- bis(dimethylamino)-2-methoxypropanenitrile in the presence of a suitable base, such as, for example piperidine, in a suitable solvent system, such as, for example, 3- methylbutan-1
  • allylpalladium chloride dimmer dichlorobis(benzonitrile)palladium (II), palladium (II) acetate, palladium (II) chloride, tetrakis(triphenylphosphine)palladium (0), tris(dibenzylideneacetone)dipalladium (0), chloro(2'-amino-1 ,1 '-biphenyl-2- yl)palladium(ll) dimer, (2'-amino-1 ,1 '-biphenyl-2-yl)methanesulfonatopalladium(ll) dimer, trans-di( ⁇ -acetato)bis[o-(di-o-tolylphosphino)benzyl]dipalladium(l l) [cataCXium® C], allylchloro[1 ,3-bis(2,4,6-trimethylphenyl)imidazol-2-ylidene
  • boronic acid or boronic acid pinacole ester of general formula (1 -8) can be reacted with a suitable boronic acid or boronic acid pinacole ester of general formula (1 -8), such as, for example (2-fluoropyridin-4-yl)boronic acid, in the presence of a suitable base, such as, for example triethylamine, a suitable activating agent such as for example N,N- dimethylpyridin-4-amine and a suitable copper salt, such as for example copper (II) acetate, in a suitable solvent system, such as, for example, trichloromethane, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at room temperature to furnish compounds of general formula (I).
  • a suitable boronic acid or boronic acid pinacole ester of general formula (1 -8)boronic acid in the presence of a suitable base, such as, for example triethylamine, a suitable activating agent such as
  • Compound (1 -9) can be converted into compounds of general formula (1 -6) by reaction with a suitable substituted acetonitlrile derivative of the general formula (1 -10), such as, for example methoxyacetonitrile, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 80 ⁇ .
  • a suitable substituted acetonitlrile derivative of the general formula (1 -10) such as, for example methoxyacetonitrile
  • Scheme 3 Process for the transformation of compounds of general formula (1 -1 1 ) into compounds of general formula (1 -13), wherein R 1 and R 2 have the meaning as given for general formula (I), supra R 12 represents a methyl- or an ethyl group or an alcohol protecting group, such as for example fert-butyldimethylsilyl, fert-butyldiphenylsilyl, triethylsilyl, triisopropylsilyl or tetrahydropyranyl and X 1 represents F, CI, Br, I or a sulfonate, e.g. trifluormethylsulfonate or p-toluolsulfonate.
  • R 1 and R 2 have the meaning as given for general formula (I)
  • R 12 represents a methyl- or an ethyl group or an alcohol protecting group, such as for example fert-butyldimethylsilyl, fert-butyldiphenylsilyl
  • Compounds of general formula (1 -1 1 ) can be converted into compounds of general formula (1 -12) by reaction with a suitable reducing agent, such as, for example borane, in a suitable solvent system, such as, for example, tetrahydrofuran, in a temperature range from - 78 ⁇ to boiling point of the respecti ve solvent, preferably the reaction is carried out at room temperature.
  • a suitable reducing agent such as, for example borane
  • a suitable solvent system such as, for example, tetrahydrofuran
  • Compounds of general formula (1 -12) can be converted into compounds of general formula (1 -13) by reaction with a suitable halogenation or sulfonylation agent, such as for example hydrogen bromide, in a suitable solvent, such as, for example, acetic acid, in a temperature range from 0 "C to the boiling poi nt of the respective solvent, preferably the reaction is carried out at room temperature.
  • a suitable halogenation or sulfonylation agent such as for example hydrogen bromide
  • a suitable solvent such as, for example, acetic acid
  • Scheme 4 Process for the transformation of compounds of general formula (1 -14) into compounds of general formula (1 -13), wherein R 1 and R 2 have the meaning as given for general formula (I), R 12 represents a methyl- or an ethyl group or an alcohol protecting group, such as for example fert-butyldimethylsilyl, fert-butyldiphenylsilyl, triethylsilyl, triisopropylsilyl or tetrahydropyranyl, X 1 and X 3 represent F, CI, Br, I or a sulfonate, e.g. trifluormethylsulfonate or p-toluolsulfonate.
  • R 1 and R 2 have the meaning as given for general formula (I)
  • R 12 represents a methyl- or an ethyl group or an alcohol protecting group, such as for example fert-butyldimethylsilyl, fert-butyldiphenylsily
  • Compounds of general formula (1 -12) can be converted into compounds of general formula (1 -13) by reaction with a suitable halogenation or sulfonylation agent, such as for example hydrogen bromide, in a suitable solvent, such as, for example, acetic acid, in a temperature range from 0 "C to the boiling poi nt of the respective solvent, preferably the reaction is carried out at room temperature.
  • a suitable halogenation or sulfonylation agent such as for example hydrogen bromide
  • a suitable solvent such as, for example, acetic acid
  • Scheme 5 Route for the preparation of compounds of general formula (la), wherein R 1 , R 2 , R 4 , R 7 , R 8 , V, W, Y and Z have the meaning as given for general formula (I), supra, R 12 represents a methyl- or an ethyl group or an alcohol protecting group, such as for example fert-butyldimethylsilyl, fert-butyldiphenylsilyl, triethylsilyl, triisopropylsilyl or tetrahydropyranyl, X 1 represents F, CI, Br, I or a sulfonate, e.g.
  • X 2 represents F, CI, Br, I, boronic acid or a boronic acid ester, such as for example 4, 4, 5, 5-tetramethyl-2-phenyl-1 ,3,2-dioxaborolane (boronic acid pinacole ester).
  • interconversion of any of the substituents R 1 , R 2 , R 4 , R 7 , R 8 , V, W, Y or Z can be achieved before and/or after the exemplified transformations.
  • modifications can be such as the introduction of protecting groups, cleavage of protecting groups, reduction or oxidation of functional groups, halogenation, metallation, substitution or other reactions known to the person skilled in the art.
  • 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.
  • Intermediates of general formula (1 -17) can be converted to intermediates of general formula (1 -18) by reaction with a suitably substituted 3,3-bis- (dimethylamino)propanenitrile of the general formula (1 -6), such as, for example 3,3- bis(dimethylamino)-2-methoxypropanenitrile, in the presence of a suitable base, such as, for example piperidine, in a suitable solvent system, such as, for example, 3- methylbutan-1 -ol, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 100 ⁇ .
  • a suitably substituted 3,3-bis- (dimethylamino)propanenitrile of the general formula (1 -6), such as, for example 3,3- bis(dimethylamino)-2-methoxypropanenitrile in the presence of a suitable base, such as, for example piperidine, in a suitable solvent system, such as, for example, 3- methylbutan-1
  • allylpalladium chloride dimmer dichlorobis(benzonitrile)palladium (II), palladium (II) acetate, palladium (II) chloride, tetrakis(triphenylphosphine)palladium (0), tris(dibenzylideneacetone)dipalladium (0), chloro(2'-amino-1 ,1 '-biphenyl-2- yl)palladium(ll) dimer, (2'-amino-1 ,1 '-biphenyl-2-yl)methanesulfonatopalladium(ll) dimer, trans-di( ⁇ -acetato)bis[o-(di-o-tolylphosphino)benzyl]dipalladium(ll) [cataCXium® C], allylchloro[1 ,3-bis(2,4,6-trimethylphenyl)imidazol-2-ylidene]pal
  • boronic acid or boronic acid pinacole ester of general formula (1 -8) can be reacted with a suitable boronic acid or boronic acid pinacole ester of general formula (1 -8), such as, for example (2-fluoropyridin-4-yl)boronic acid, in the presence of a suitable base, such as, for example triethylamine, a suitable activating agent such as for example N,N- dimethylpyridin-4-amine and a suitable copper salt, such as for example copper (II) acetate, in a suitable solvent system, such as, for example, trichloromethane, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at room temperature to furnish compounds of general formula (1 -19).
  • a suitable boronic acid or boronic acid pinacole ester of general formula (1 -8)boronic acid in the presence of a suitable base, such as, for example triethylamine, a suitable activating agent such
  • intermediates of general formula (1 -18) can be reacted with a suitable six membered heterocycle of the general formula (1 -8), such as for example 4-fluoro-2- methyl-pyridine, in the presence of a suitable base, such as, for example sodium hydride, in a suitable solvent system, such as, for example DMF, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 90 ⁇ to furnish com pounds of general formula (1 -19).
  • a suitable six membered heterocycle of the general formula (1 -8) such as for example 4-fluoro-2- methyl-pyridine
  • a suitable base such as, for example sodium hydride
  • a suitable solvent system such as, for example DMF
  • Compounds of general formula (1 -19) are converted to compounds of general formula (la) by treatment with a suitable deprotection agent, such as a dealkylating agent, such as for example boron trichloride, in a suitable solvent, such as, for example, dichloromethane, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at room temperature.
  • a suitable deprotection agent such as a dealkylating agent, such as for example boron trichloride
  • a suitable solvent such as, for example, dichloromethane
  • any of the substituents R 1 , R 2 , R 3 or R 4 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, 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, 3 rd edition, Wiley 1999). Specific examples are described in the subsequent para-graphs.
  • Intermediates of general formula (1 -5) can be converted to intermediates of general formula (1 -7) by reaction with a suitably substituted 3-methoxyacrylonitrile of the general formula (1 -20), such as, for example (ethoxymethylene)malononitrile, in the presence of a suitable base, such as, for example sodium methanolate, in a suitable solvent system, such as, for example, methanol, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 65 ⁇ .
  • a suitably substituted 3-methoxyacrylonitrile of the general formula (1 -20) such as, for example (ethoxymethylene)malononitrile
  • a suitable base such as, for example sodium methanolate
  • a suitable solvent system such as, for example, methanol
  • R 9 represents a methyl- or an ethyl group or an alcohol protecting group, such as for example tert- butyldimethylsilyl, tert-butyldiphenylsilyl, triethylsilyl, triisopropylsilyl or tetrahydropyranyl.
  • Intermediates of general formula (1 -4) can be converted to intermediates of general formula (1 -21 ) by reaction with ammonia, in a suitable solvent system, such as, for example, methanol, at a temperature between 0 ⁇ an d boiling point of the respective solvent, preferably the reaction is carried out at 50 ⁇ , at a pressure between 1 and 10 bar, preferably the reaction is carried in a sealed vessel.
  • a suitable solvent system such as, for example, methanol
  • Intermediates of general formula (1 -22) can be converted to intermediates of general formula (1 -5) by reaction with a suitable alcoholate, such as, for example sodium methanolate, in a suitable solvent system, such as, for example, the corresponding alcohol, e.g. methanol, at a temperature between room temperature and the boiling point of the respective solvent, preferably the reaction is carried out at room temperature, and subsequent treatment with a suitable source of ammonium, such as for example, ammonium chloride in the presence of a suitable acid, such as for example acetic acid in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 50 ⁇ .
  • a suitable alcoholate such as, for example sodium methanolate
  • a suitable solvent system such as, for example, the corresponding alcohol, e.g. methanol
  • Scheme 8 Route for the preparation of compounds of general formula (1 -22), R 1 , R 2 , R 3 , R 7 and R 8 have the meaning as given for general formula (I), supra, X 1 represents F, CI, Br, I or a sulfonate, e.g. trifluormethylsulfonate or p-toluolsulfonate.
  • Compounds of general formula (1 -29) are either commercially available (i.e. CAS Reg. Nos. 68618-36-0, 76006-13-8, 633328-33-3, 1785345-95-0, 1351386-47-4, 1781061 - 48-0, 1780665-28-2), or can be prepared according to procedures available from the public domain, as understandable to the person skilled in the art. Specific examples are described in the subsequent paragraphs.
  • Intermediates of general formula (1 -29) can be converted to intermediates of general formula (1 -30) by reaction with a suitable alkylating agent, such as, for example a substituted benzyl halide (1 -3), in the presence of a suitable base, such as, for example cesium carbonate or sodium hydride, in a suitable solvent system, such as, for example, DMF, at a temperature between - 20 ⁇ and boiling point of the respective solvent, preferably the reaction is carried out at 0 ⁇ .
  • a suitable alkylating agent such as, for example a substituted benzyl halide (1 -3
  • a suitable base such as, for example cesium carbonate or sodium hydride
  • a suitable solvent system such as, for example, DMF
  • Intermediates of general formula (1 -30) can be converted to intermediates of general formula (1 -22) by reaction with zinc cynanide, in the presence of a catalyst, such as for example [1 ,1 '-bis(diphenylphosphino)ferrocene]dichloropalladium(ll) complex with dichloromethane, in a suitable solvent system, such as, for example, DMF, at a temperature between 50 ⁇ and the boiling point of t he respective solvent, preferably the reaction is carried out at 120 ⁇ .
  • a catalyst such as for example [1 ,1 '-bis(diphenylphosphino)ferrocene]dichloropalladium(ll) complex with dichloromethane
  • a suitable solvent system such as, for example, DMF
  • Scheme 9 Route for the preparation of compounds of general formula (I), wherein R 1 , R 2 , R 3 , R 4 , V, W, Y and Z have the meaning as given for general formula (I), supra, X 1 represents F, CI, Br, I or a sulfonate, e.g. trifluormethylsulfonate or p-toluolsulfonate, and X 2 represents F, CI, Br, I, boronic acid or a boronic acid ester, such as for example 4,4,5,5-tetramethyl-2-phenyl-1 ,3,2-dioxaborolane (boronic acid pinacole ester).
  • X 1 represents F, CI, Br, I or a sulfonate, e.g. trifluormethylsulfonate or p-toluolsulfonate
  • X 2 represents F, CI, Br, I, boronic acid or a boronic acid ester,
  • interconversion of any of the substituents R 1 , R 2 , R 3 , R 4 , V, W, Y or Z can be achieved before and/or after the exemplified transformations.
  • modifications can be such as the introduction of protecting groups, cleavage of protecting groups, reduction or oxidation of functional groups, halogenation, metallation, substitution or other reactions known to the person skilled in the art.
  • 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, 3 rd edition, Wiley 1999). Specific examples are described in the subsequent paragraphs.
  • Intermediates of general formula (1 -25) can be converted to intermediates of general formula (1 -26) by reaction with a suitably substituted 3,3-bis- (dimethylamino)propanenitrile of the general formula (1 -6), such as, for example 3,3- bis(dimethylamino)-2-methoxypropanenitrile, in the presence of a suitable base, such as, for example piperidine, in a suitable solvent system, such as, for example, 3- methylbutan-1 -ol, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 100 ⁇ .
  • a suitably substituted 3,3-bis- (dimethylamino)propanenitrile of the general formula (1 -6), such as, for example 3,3- bis(dimethylamino)-2-methoxypropanenitrile in the presence of a suitable base, such as, for example piperidine, in a suitable solvent system, such as, for example, 3- methylbutan-1
  • Intermediates of general formula (1 -26) can be converted to intermediates of general formula (1 -27) by reaction with a suitably Broensted acid, such as, for example methanesulfonic acid and trifluoroacetic acid, in a suitable solvent system, such as, for example, dichloromethane, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at room temperature.
  • a suitably Broensted acid such as, for example methanesulfonic acid and trifluoroacetic acid
  • a suitable solvent system such as, for example, dichloromethane
  • Intermediates of the general formula (1 -27) can be converted to intermediates of general formula (1 -7) by reaction with a suitable alkylating agent, such as, for example a substituted benzyl halide (1 -3), in the presence of a suitable base, such as, for example sodium hydride, in a suitable solvent system, such as, for example, DMF, at a temperature between - 20 ⁇ and boiling point of th e respective solvent, preferably the reaction is carried out at 0 ⁇ .
  • a suitable alkylating agent such as, for example a substituted benzyl halide (1 -3
  • a suitable base such as, for example sodium hydride
  • a suitable solvent system such as, for example, DMF
  • allylpalladium chloride dimmer dichlorobis(benzonitrile)palladium (II), palladium (II) acetate, palladium (II) chloride, tetrakis(triphenylphosphine)palladium (0), tris(dibenzylideneacetone)dipalladium (0), chloro(2'-amino-1 ,1 '-biphenyl-2- yl)palladium(ll) dimer, (2'-amino-1 ,1 '-biphenyl-2-yl)methanesulfonatopalladium(ll) dimer, trans-di( ⁇ -acetato)bis[o-(di-o-tolylphosphino)benzyl]dipalladium(ll) [cataCXium® C], allylchloro[1 ,3-bis(2,4,6-trimethylphenyl)imidazol-2-ylidene]pal
  • boronic acid or boronic acid pinacole ester of general formula (1 -8) can be reacted with a suitable boronic acid or boronic acid pinacole ester of general formula (1 -8), such as, for example (2-fluoropyridin-4-yl)boronic acid, in the presence of a suitable base, such as, for example triethylamine, a suitable activating agent such as for example N,N- dimethylpyridin-4-amine and a suitable copper salt, such as for example copper (II) acetate, in a suitable solvent system, such as, for example, trichloromethane, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at room temperature to furnish compounds of general formula (I).
  • a suitable boronic acid or boronic acid pinacole ester of general formula (1 -8)boronic acid in the presence of a suitable base, such as, for example triethylamine, a suitable activating agent such as
  • the compounds according to the invention are isolated and purified in a manner known per se, e.g. by distilling off the solvent in vacuo and recrystallizing the residue obtained from a suitable solvent or subjecting it to one of the customary purification methods, such as chromatography on a suitable support material.
  • reverse phase preparative HPLC of compounds of the present invention which possess a sufficiently basic or acidic functionality may result in the formation of a salt, such as, in the case of a compound of the present invention which is sufficiently basic, a trifluoroacetate or formate salt for example, or, in the case of a compound of the present invention which is sufficiently acidic, an ammonium salt for example.
  • Salts of this type can either be transformed into its free base or free acid form, respectively, by various methods known to the persion skilled in the art, or be used as salts in subsequent biological assays. Additionally, the drying process during the isolation of compounds of the present invention may not fully remove traces of cosolvents, especially such as formic acid or trifluoroacetic acid, to give solvates or inclusion complexes. The person skilled in the art will recognise which solvates or inclusion complexes are acceptable to be used in subsequent biological assays. It is to be understood that the specific form (e.g. salt, free base, solvate, inclusion complex) of a compound of the present invention as isolated as described herein is not necessarily the only form in which said compound can be applied to a biological assay in order to quantify the specific biological activity.
  • Salts of the compounds of formula (I) according to the invention can be obtained by dissolving the free compound in a suitable solvent (for example a ketone such as acetone, methylethylketone or methylisobutylketone, an ether such as diethyl ether, tetrahydrofuran or dioxane, a chlorinated hydrocarbon such as methylene chloride or chloroform, or a low molecular weight aliphatic alcohol such as methanol, ethanol or isopropanol) which contains the desired acid or base, or to which the desired acid or base is then added.
  • a suitable solvent for example a ketone such as acetone, methylethylketone or methylisobutylketone, an ether such as diethyl ether, tetrahydrofuran or dioxane, a chlorinated hydrocarbon such as methylene chloride or chloroform, or a low molecular weight aliphatic alcohol
  • the acid or base can be employed in salt preparation, depending on whether a mono- or polybasic acid or base is concerned and depending on which salt is desired, in an equimolar quantitative ratio or one differing therefrom.
  • the salts are obtained by filtering, reprecipitating, precipitating with a non-solvent for the salt or by evaporating the solvent. Salts obtained can be converted into the free compounds which, in turn, can be converted into salts.
  • pharmaceutically unacceptable salts which can be obtained, for example, as process products in the manufacturing on an industrial scale, can be converted into pharmaceutically acceptable salts by processes known to the person skilled in the art.
  • hydrochlorides and the process used in the examples section are especially preferred.
  • Pure diastereomers and pure enantiomers of the compounds and salts according to the invention can be obtained e.g. by asymmetric synthesis, by using chiral starting compounds in synthesis and by splitting up enantiomeric and diasteriomeric mixtures obtained in synthesis.
  • Enantiomeric and diastereomeric mixtures can be split up into the pure enantiomers and pure diastereomers by methods known to a person skilled in the art. Preferably, diastereomeric mixtures are separated by crystallization, in particular fractional crystallization, or chromatography. Enantiomeric mixtures can be separated e.g. by forming diastereomers with a chiral auxiliary agent, resolving the diastereomers obtained and removing the chiral auxiliary agent.
  • chiral auxiliary agents for example, chiral acids can be used to separate enantiomeric bases such as e.g. mandelic acid and chiral bases can be used to separate enantiomeric acids via formation of diastereomeric salts.
  • diastereomeric derivatives such as diastereomeric esters can be formed from enantiomeric mixtures of alcohols or enantiomeric mixtures of acids, respectively, using chiral acids or chiral alcohols, respectively, as chiral auxiliary agents.
  • diastereomeric complexes or diastereomeric clathrates may be used for separating enantiomeric mixtures.
  • enantiomeric mixtures can be split up using chiral separating columns in chromatography. Another suitable method for the isolation of enantiomers is the enzymatic separation.
  • One preferred aspect of the invention is the process for the preparation of the compounds of claims 1 -6 according to the examples.
  • compounds of the formula (I) can be converted into their salts, or, optionally, salts of the compounds of the formula (I) can be converted into the free compounds.
  • Corresponding processes are customary for the skilled person.
  • N-oxides can be converted into their N-oxides.
  • the N- oxide may also be introduced by way of an intermediate.
  • N-oxides may be prepared by treating an appropriate precursor with an oxidizing agent, such as meta- chloroperbenzoic acid, in an appropriate solvent, such as DCM, at suitable temperatures, such as from 0 ⁇ to 40 "C, whereby r oom temperature is generally preferred. Further corresponding processes for forming N-oxides are customary for the skilled person.
  • One preferred aspect of the invention is the process for the preparation of the compounds of claims 1 -6 according to the examples, as well as the intermediates used for their preparation.
  • compounds of the formula (I) can be converted into their salts, or, optionally, salts of the compounds of the formula (I) can be converted into the free compounds.
  • Corresponding processes are customary for the skilled person.
  • the compounds of the present invention have surprisingly been found to effectively inhibit Bub1 finally resulting in cell death e.g. apoptosis 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 is mediated by Bub1 , such as, for example, benign and malignant neoplasia, more specifically 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,
  • Haematological tumors can e.g be exemplified by aggressive and indolent forms of leukemia and lymphoma, namely non-Hodgkins disease, chronic and acute myeloid leukemia (CML / AML), acute lymphoblastic leukemia (ALL), Hodgkins disease, multiple myeloma and T-cell lymphoma. Also included are myelodysplastic syndrome, plasma cell neoplasia, paraneoplastic syndromes, and cancers of unknown primary site as well as AIDS related malignancies.
  • a further aspect of the invention is the use of the compounds according to formula (I) for the treatment of cer-vical -, breast -, non-small cell lung -, prostate -, colon - and melanoma tumors and/or metastases thereof, especially preferred for the treatment thereof as well as a method of treatment of cervical -, breast -, non-small cell lung -, prostate -, colon - and melanoma tumors and/or metastases thereof comprising administering an effective amount of a compound of formula (I).
  • One aspect of the invention is the use of the compounds according to formula (I) for the treatment of cervix tumors as well as a method of treatment of cervix tumors comprising administering an effective amount of a compound of formula (I).
  • the invention relates to a compound of general formula 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 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, especially for use in the 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 the prophylaxis or treatment of hyperproliferative disorders or disorders responsive to induction of cell death i.e apoptosis. .
  • 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 preferably 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, especially the treatment, wherein the diseases are haematological tumours, solid tumours and/or metastases thereof.
  • Another aspect is the use of a compound of formula (I) is for the treatment of cervical -, breast -, non-small cell lung -, prostate -, colon - and melanoma tumors and/or metastases thereof, especially preferred for the treatment thereof.
  • a preferred aspect is the use of a compound of formula (I) for the prophylaxis and/or treatment of cervical tumors especially preferred for the treatment thereof.
  • 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, wherein such disease is a hyperproliferative disorder or a disorder responsive to induction of cell death e.g.apoptosis.
  • the disease is a haematological tumour, a solid tumour and/or metastases thereof.
  • the disease is cervical -, breast -, non-small cell lung -, prostate -, colon - and melanoma tumor and/or metastases thereof, in a preferred aspect the disease is cervical tumor.
  • 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 cell death e.g. 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.
  • Hyper-proliferative 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.
  • 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 AIDS-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.
  • Methods of treating kinase disorders The present invention also provides methods for the treatment of disorders associated with aberrant mitogen extracellular kinase activity, including, but not limited to stroke, heart failure, hepatomegaly, cardiomegaly, diabetes, Alzheimer's disease, cystic fibrosis, symptoms of xenograft rejections, septic shock or asthma.
  • Effective amounts of compounds of the present invention can be used to treat such disorders, including those diseases (e.g., cancer) mentioned in the Background section above. Nonetheless, such cancers and other diseases can be treated with compounds of the present invention, regardless of the mechanism of action and/or the relationship between the kinase and the disorder.
  • aberrant kinase activity or "aberrant tyrosine kinase activity,” includes any abnormal expression or activity of the gene encoding the kinase or of the polypeptide it encodes. Examples of such aberrant activity, include, but are not limited to, over- expression of the gene or polypeptide ; gene amplification ; mutations which produce constitutively-active or hyperactive kinase activity ; gene mutations, deletions, substitutions, additions, etc.
  • the present invention also provides for methods of inhibiting a kinase activity, especially of mitogen extracellular kinase, comprising administering an effective amount of a compound of the present invention, including salts, polymorphs, metabolites, hydrates, solvates, prodrugs (e.g.: esters) thereof, and diastereoisomeric forms thereof.
  • Kinase activity can be inhibited in cells (e.g., in vitro), or in the cells of a mammalian subject, especially a human patient in need of treatment.
  • 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 , 1480 ; 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
  • restenosis in-stent restenosis
  • vascular graft restenosis etc.
  • 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 e.g. apoptosis of such cell types.
  • 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, especially in therapy of tumour growth and metastases, especially in solid tumours of all indications and stages with or without pre-treatment of the tumour growth.
  • compositions of the compounds of the invention are provided.
  • 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.
  • the present invention includes pharmaceutical compositions that are comprised of a pharmaceutically acceptable carrier or auxiliary and a pharmaceutically effective amount of a compound, or salt thereof, of the present invention.
  • Another aspect of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically effective amount of a compound of formula (I) and a pharmaceutically acceptable auxiliary for the treatment of a disease mentioned supra, especially for the treatment of haematological tumours, solid tumours and/or metastases thereof.
  • a pharmaceutically acceptable carrier or auxiliary is preferably a carrier that is 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.
  • Carriers and auxiliaries are all kinds of additives assisting to the composition to be suitable for administration.
  • a pharmaceutically effective amount of compound is preferably that amount which produces a result or exerts the intended influence on the particular condition being treated.
  • the compounds of the present invention can be administered with pharmaceutically- acceptable carriers or auxiliaries 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 auxiliaries, 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.
  • the suspensions may also contain one or more preservatives, for example, ethyl or n-propyl p-hydroxybenzoate ; 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 poly(ethylene 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 sur
  • 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 quarternary ammonium salts, as well as mixtures.
  • suitable detergents include cationic detergents, for example
  • 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 a
  • 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.
  • Controlled release formulations for parenteral administration include liposomal, polymeric microsphere and polymeric gel formulations that are known in the art.
  • 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.
  • 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) ; alkalinizing 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 charcoa)l ; aerosol propellants (examples include but are not limited to carbon dioxide, CCI 2 F 2 , 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, propy
  • 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 include but are not limited to bentonite
  • emulsifying agents include but are not limited to acacia, cetomacrogol, cetyl alcohol, glyceryl monostearate, lecithin, sorbitan monooleate, polyoxyethylene 50 monostearate
  • encapsulating agents 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 include but are not limited to glycerol, propylene glycol and sorbitol
  • compositions according to the present invention can be illustrated as follows:
  • Sterile i.v. 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 i.v. infusion over about 60 minutes.
  • 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 minutes.
  • 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.
  • a digestible oil such as soybean oil, cottonseed oil or olive oil is prepared and injected by means of a positive displacement pump
  • 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.
  • 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 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.
  • Those combined pharmaceutical agents can be other agents having antiproliferative effects such as for example for the treatment of haematological tumours, solid tumours and/or metastases thereof and/or agents for the treatment of undesired side effects.
  • the present invention relates also to such combinations.
  • anti-hyper-proliferative agents suitable for use with the composition of the invention include but are not limited to those compounds acknowledged to be used in the treatment of neoplastic diseases in Goodman and Gilman's The Pharmacological Basis of Therapeutics (Ninth Edition), editor Molinoff et al., publ. by McGraw-Hill, pages 1225-1287, (1996), which is hereby incorporated by reference, especially (chemotherapeutic) anti-cancer agents as defined supra.
  • the combination can be a non-fixed combination or a fixed-dose combination as the case may be. Methods of testing for a particular pharmacological or pharmaceutical property are well known to persons skilled in the art.
  • NMR nuclear magnetic resonance spectroscopy chemical shifts ( ⁇ ) are given in ppm. The chemical shifts were corrected by setting the DMSO signal to 2.50 ppm using unless otherwise stated.
  • PoraPakTM a HPLC column obtainable from Waters
  • NMR peak forms in the following specific experimental descriptions are stated as they appear in the spectra, possible higher order effects have not been considered.
  • Reactions employing microwave irradiation may be run with a Biotage Initator® microwave oven optionally equipped with a robotic unit.
  • the reported reaction times employing microwave heating are intended to be understood as fixed reaction times after reaching the indicated reaction temperature.
  • the compounds and intermediates produced according to the methods of the invention may require purification. Purification of organic compounds is well known to the person skilled in the art and there may be several ways of purifying the same compound. In some cases, no purification may be necessary. In some cases, the compounds may be purified by crystallization. In some cases, impurities may be stirred out using a suitable solvent.
  • the compounds may be purified by chromatography, particularly flash column chromatography, using for example prepacked silica gel cartridges, e.g. from Separtis such as Isolute® Flash silica gel or Isolute® Flash NH 2 silica gel in combination with a Isolera® autopurifier (Biotage) and eluents such as gradients of e.g. hexane/ethyl acetate or DCM/methanol.
  • Separtis such as Isolute® Flash silica gel or Isolute® Flash NH 2 silica gel in combination with a Isolera® autopurifier (Biotage) and eluents such as gradients of e.g. hexane/ethyl acetate or DCM/methanol.
  • the compounds may be purified by preparative HPLC using for example a Waters autopurifier equipped with a diode array detector and/or on-line electrospray ionization mass spectrometer in combination with a suitable prepacked reverse phase column and eluents such as gradients of water and acetonitrile which may contain additives such as trifluoroacetic acid, formic acid or aqueous ammonia.
  • a Waters autopurifier equipped with a diode array detector and/or on-line electrospray ionization mass spectrometer in combination with a suitable prepacked reverse phase column and eluents such as gradients of water and acetonitrile which may contain additives such as trifluoroacetic acid, formic acid or aqueous ammonia.
  • purification methods as described above can provide those compounds of the present invention which possess a sufficiently basic or acidic functionality in the form of a salt, such as, in the case of a compound of the present invention which is sufficiently basic, a trifluoroacetate or formate salt for example, or, in the case of a compound of the present invention which is sufficiently acidic, an ammonium salt for example.
  • a salt of this type can either be transformed into its free base or free acid form, respectively, by various methods known to the person skilled in the art, or be used as salts in subsequent biological assays. It is to be understood that the specific form (e.g. salt, free base etc) of a compound of the present invention as isolated as described herein is not necessarily the only form in which said compound can be applied to a biological assay in order to quantify the specific biological activity.
  • A1 water + 0.1 % vol. formic acid (99%)
  • the crude product was purified by flash chromatography.
  • the product fraction was treated with ethyl acetate and strirred for a few minutes.
  • the unsolved precipitate was filtered off, washed with ethyl acetate and dried at 5CC under vacuo. The fi ltrate was dried under reduced pressure.
  • the filtrate was treated with methanol and strirred for a few minutes.
  • the unsolved precipitate was filtered off, washed with methanol.
  • the filtrate was concentrated under reduced pressure.
  • the filtrate and both residues were purified by flash chromatography to provide 6.2 g of the 75% pure target compound: 12.6 mmol,
  • the reaction mixture was concentrated under reduced pressure and purified by flash chromatography and HPLC to provide the target compound as salt in 100% purity: 260 mg. It was dissolved in methylene chloride and a few drops methanol and treated with saturated sodium hydogen carbonate solution. It was stirred for 5 minutes and the organic layer was filtered through a silicone coated filter and concentrated under reduced pressure: 237 mg, 100% pure, 14 % yield, 0.54 mmol.
  • the reaction mixture was diluted with 10 mL methanol. The mixture was stirred at 50 ⁇ overnight. Under heating the r eaction mixture turned clear. The reaction mixture was concentrated under vacuo. The residue was diluted with dichloromethane and 1 M hydrochloric acid. The layers were separated and the aqueous layer was extracted with dichloromethane twice. The combined organic layers were dried using a waterresistant filter and concentrated under reduced pressure. The aqueous layer was concentrated under reduced pressure to provide the target compound with 90% purity: 4.12 g, 10.1 mmol, 106%.
  • the reactio n mixture was treated with dichloromethane and water and stirred for a few minutes.
  • the organic layer was filtered through a silicone coated filter and concentrated under reduced pressure.
  • the crude product was treated with methanol and the unsolved precipitate was filtered off, washed with methanol and dried at 50 ⁇ under vacuo: 88 mg, 100% pure, 62% yield, 0.16 mmol.
  • reaction mixture was quenshed with ice water and the pH was adjusted to 10 by addition of sodium hydroxide solution (2 M). It was extracted 3 times with dichloromethane / isopropanole 9:1 , washed once with water and brine, filtered through a silicone coated filter and concentrated in vacuo.
  • the crude product was purified by HPLC chromatography under basic conditions: 21 mg, 95% pure, 26% yield, 37 ⁇ .
  • the following assays can be used to illustrate the commercial utility of the compounds according to the present invention. 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
  • 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 calculated utilizing data sets obtained from testing of one or more synthetic batch.
  • Bub1 -inhibitory activities of compounds described in the present invention were quantified using a time-resolved fluorescence energy transfer (TR-FRET) kinase assay which measures phosphorylation of the synthetic peptide Biotin-Ahx-VLLPKKSFAEPG (C-terminus in amide form), purchased from e.g. Biosyntan (Berlin, Germany) by the (recombinant) catalytic domain of human Bub1 (amino acids 704-1085), expressed in Hi5 insect cells with an N-terminal His6-tag and purified by affinity- (Ni-NTA) and size exclusion chromatography.
  • TR-FRET time-resolved fluorescence energy transfer
  • Bub1 the final concentration of Bub1 was adjusted depending on the activity of the enzyme lot in order to be within the linear dynamic range of the assay: typically ⁇ 200 ng/mL were used
  • aqueous assay buffer [50 mM Tris/HCI pH 7.5, 10 mM magnesium chloride (MgCI 2 ), 200 mM potassium chloride (KCI), 1 .0 mM dithiothreitol (DTT), 0.1 mM sodium ortho-vanadate, 1 % (v/v) glycerol, 0.01 % (w/v) bovine serum albumine (BSA), 0.005% (v/v) Trition X-100 (Sigma), 1 x Complete EDTA-free protease inhibitor mixture (Roche)] were added to the compounds in the test plate and the mixture was incubated for 15 min at 22 ⁇ to allow pre-equilibration of th e putative enzyme-inhibitor complexes before the start of
  • the resulting mixture (5 ⁇ _ final volume) was incubated at 22 ⁇ d uring 60 min., and the reaction was stopped by the addition of 5 ⁇ _ of an aqueous EDTA-solution (50 mM EDTA, in 100 mM HEPES pH 7.5 and 0.2 % (w/v) bovine serum albumin) which also contained the TR- FRET detection reagents (0.2 ⁇ streptavidin-XL665 [Cisbio Bioassays, Codolet, France] and 1 nM anti-phosho-Serine antibody [Merck Millipore, cat. # 35-001 ] and 0.4 nM LANCE EU-W1024 labeled anti-mouse IgG antibody [Perkin-Elmer, product no.
  • TR- FRET detection reagents 0.2 ⁇ streptavidin-XL665 [Cisbio Bioassays, Codolet, France] and 1 nM anti-phosho-Serine antibody [Merck Millipore, cat
  • AD0077 alternatively a Terbium -cryptate-labeled anti-mouse IgG antibody from Cisbio Bioassays can be used]).
  • the stopped reaction mixture was further incubated 1 h at 22 ⁇ in order to allow the formation of complexes b etween peptides and detection reagents.
  • the amount of product was evaluated by measurement of the resonance energy transfer from the Eu-chelate-antibody complex recognizing the Phosphoserine residue to the streptavidin-XL665 bound to the biotin moiety of the peptide.
  • the fluorescence emissions at 620 nm and 665 nm after excitation at 330-350 nm were measured in a TR-FRET plate reader, e.g. a Rubystar or Pherastar (both from BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin-Elmer) and the ratio of the emissions (665 nm/622 nm) was taken as indicator for the amount of phosphorylated substrate.
  • TR-FRET plate reader e.g. a Rubystar or Pherastar (both from BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin-Elmer) and the ratio of the emissions (665 nm/622 nm) was taken as indicator for the amount of phosphorylated substrate.
  • the cells of one plate were stained with crystal violet (see below), while the medium of the other plates was replaced by fresh medium containing the test substances in various concentrations (0 ⁇ , as well as in the range of 0.001 -10 ⁇ ; the final concentration of the solvent dimethyl sulfoxide was 0.5%).
  • the cells were incubated for 4 days in the presence of test substances.
  • Cell proliferation was determined by staining the cells with crystal violet: the cells were fixed by adding 20 ⁇ -, ⁇ ⁇ point of an 1 1 % glutaric aldehyde solution for 15 minutes at room temperature. After three washing cycles of the fixed cells with water, the plates were dried at room temperature.
  • the cells of one plate were stained with crystal violet (see below), while the medium of the other plates was supplemented with the test substances in various concentrations (0 ⁇ , as well as in the range of 0.001 -10 ⁇ ; the final concentration of the solvent dimethyl sulfoxide was adjusted to 0.1 %) using a Hewlett-Packard HP D300 Digital Dispenser.
  • the cells were incubated for 4 days in the presence of test substances.
  • Cell proliferation was determined by staining the cells with crystal violet: the cells were fixed by adding 20 ⁇ -ymeasuring point of an 11 % glutaric aldehyde solution for 15 minutes at room temperature. After three washing cycles of the fixed cells with water, the plates were dried at room temperature.
  • Cultivated HeLa human cervical tumor cells (DSMZ ACC-57) were plated at a density of 3000 cells/well in a 96-well multititer plate in 100 ⁇ _ of growth medium supplemented 10% fetal calf serum. After 24 hours, the cells of one plate (zero-point plate) were stained with crystal violet (see below), while the medium of the other plates was replaced by fresh culture medium (100 ⁇ _) containing 3 nM of paclitaxel (Sigma- Aldrich).
  • Cultivated HeLa human cervical tumor cells (DSMZ ACC-57) were plated at a density of 3000 cells/well in a 96-well multititer plate in 100 ⁇ _ of growth medium supplemented 10% fetal calf serum. After 24 hours, the cells of one plate (zero-point plate) were stained with crystal violet (see below), while the medium of the other plates was replaced by fresh culture medium containing 3 nM of paclitaxel (Sigma-Aldrich).
  • test substances were a dded in various concentrations (0 ⁇ , as well as in the range of 0.001 -10 ⁇ ; the final concentration of the solvent dimethyl sulfoxide was adjusted to 0.1 %) using a Hewlett-Packard HP D300 Digital Dispenser.
  • the cells were incubated for another 92 hours at 37 ⁇ in the presence of test substances.
  • Cell proliferation was determined by staining the cells with crystal violet: the cells were fixed by adding 20 ⁇ point of an 1 1 % glutaric aldehyde solution for 15 minutes at room temperature. After three washing cycles of the fixed cells with water, the plates were dried at room temperature. The cells were stained by adding 100 ⁇ point of a 0.1 % crystal violet solution (pH 3.0).
  • the plates were dried at room temperature.
  • the IC 5 o values were determined by means of a 4 parameter fit.
  • This assay determines the suppression of histone 2A phosphorylation by a Bub1 kinase inhibitor during co-treatment with Nocodazole.
  • 25000 cells (cells were ordered from ATCC) are seeded in 96well plate for 5 h at 37 ⁇ . C ells are treated with Nocodazole ( ⁇ g/ml) and varying concentrations (between 3nM and 10 ⁇ ) of test compounds for 16h. Cells are fixed (20 min, Fixing solution R&D), washed three times with PBS and blocked with Odyssey blocking buffer before incubating with the primary antibody against phosphorylated H2A (5 ⁇ / ⁇ ABIN482721 ) overnight at 2-8 ⁇ .
  • This assay measures the inhibition of histone 2A phosphorylation, which was induced by pre-treatment of the cells with Nocodazole, by a Bub1 kinase inhibitor.
  • 25000 cells (cells were ordered from ATCC) are seeded in 96well plate for 5 h at 37 ⁇ . Cells are treated with Nocodazole ( ⁇ g/ml). After 16h varying concentrations (between 3nM and 10 ⁇ ) of test compounds are added and the cells are incubated for another 1 h.
  • the quotient of 800nm and 700nm for Nocodazole only treated cells is set as 100% and the quotient of 800nm and 700nm of untreated cells is set as 0%.
  • the results given as % are reflecting the inhibition of Bub1 kinase activity compared to control and normalized according to cell number.
  • the IC 5 o values are determined by means of a 4 parameter fit.
  • Histone H2A is an immediate intracellular substrate of Bub1 kinase. Determination of the phosphorylation status of Histone H2A provides a direct measure of the intracellular activity of Bub1 kinase.
  • test compounds with paclitaxel in vivo 8 mg/kg of paclitaxel are injected once intravenously into the tail vein of NMRI nude mice. Immediately thereafter 50 mg/kg of the test compound is administered by gavage to mice. Blood is taken from mice following decapitation 1 , 3, 7 and 24 hours after injection of Paclitaxel. Plasma concentrations of test compound and of paclitaxel, respectively, are determined by LC/MSMS. The data from the paclitaxel mono treatment group, the test compound mono treatment group, and the combination treatment group are compared for evaluation of the drug-drug interaction potential.
  • Assay 1 Assay 2.1 : Assay 2.2: Assay 3.1 : Assay 3.2:
  • HeLa assay HeLa assay (HeLa assay (HeLa assay (HeLa mean ICso cell line) cell line) + paclitaxel) + paclitaxel)

Abstract

Compounds of formula (I) and their use as pharmaceuticals.

Description

ANNULATED PYRAZOLES AS BUB1 KINASE INHIBITORS FOR TREATING PROLIFERATIVE DISORDERS
Field of application of the invention The invention relates to substituted annulated pyrazole compounds, a process for their production and the use thereof.
BACKGROUND OF THE INVENTION One of the most fundamental characteristics of cancer cells is their ability to sustain chronic proliferation whereas in normal tissues the entry into and progression through the cell divison cycle is tightly controlled to ensure a homeostasis of cell number and maintenance of normal tissue function. Loss of proliferation control was emphasized as one of the six hallmarks of cancer [Hanahan D and Weinberg RA, Cell 100, 57, 2000; Hanahan D and Weinberg RA, Cell 144, 646, 2011 ].
The eukaryotic cell division cycle (or cell cycle) ensures the duplication of the genome and its distribution to the daughter cells by passing through a coordinated and regulated sequence of events. The cell cycle is divided into four successive phases: 1 . The G1 phase represents the time before the DNA replication, in which the cell grows and is sensitive to external stimuli.
2. In the S phase the cell replicates its DNA, and
3. in the G2 phase preparations are made for entry into mitosis.
4. In mitosis (M phase), the duplicated chromosomes get separated supported by a spindle device built from microtubules, and cell division into two daughter cells is completed.
To ensure the extraordinary high fidelity required for an accurate distribution of the chromosomes to the daughter cells, the passage through the cell cycle is strictly regulated and controlled. The enzymes that are necessary for the progression through the cycle must be activated at the correct time and are also turned off again as soon as the corresponding phase is passed. Corresponding control points ("checkpoints") stop or delay the progression through the cell cycle if DNA damage is detected, or the DNA replication or the creation of the spindle device is not yet completed. The mitotic checkpoint (also known as spindle checkpoint or spindle assembly checkpoint) controls the accurate attachment of mircrotubules of the spindle device to the kinetochors (the attachment site for microtubules) of the duplicated chromosomes. The mitotic checkpoint is active as long as unattached kinetochores are present and generates a wait-signal to give the dividing cell the time to ensure that each kinetochore is attached to a spindle pole, and to correct attachment errors. Thus the mitotic checkpoint prevents a mitotic cell from completing cell division with unattached or erroneously attached chromosomes [Suijkerbuijk SJ and Kops GJ, Biochem. Biophys. Acta 1786, 24, 2008; Musacchio A and Salmon ED, Nat. Rev. Mol. Cell. Biol. 8, 379, 2007]. Once all kinetochores are attached with the mitotic spindle poles in a correct bipolar (amphitelic) fashion, the checkpoint is satisfied and the cell enters anaphase and proceeds through mitosis.
The mitotic checkpoint is established by a complex network of a number of essential proteins, including members of the MAD (mitotic arrest deficient, MAD 1 -3) and Bub (Budding uninhibited by benzimidazole, Bub 1 -3) families, Mps1 kinase, cdc20, as well as other components [reviewed in Bolanos-Garcia VM and Blundell TL, Trends Biochem. Sci. 36, 141 , 2010], many of these being over-expressed in proliferating cells (e.g. cancer cells) and tissues [Yuan B et al., Clin. Cancer Res. 12, 405, 2006]. The major function of an unsatisfied mitotic checkpoint is to keep the anaphase-promoting complex/cyclosome (APC/C) in an inactive state. As soon as the checkpoint gets satisfied the APC/C ubiquitin-ligase targets cyclin B and securin for proteolytic degradation leading to separation of the paired chromosomes and exit from mitosis.
Inactive mutations of the Ser/Thr kinase Bub1 prevented the delay in progression through mitosis upon treatment of cells of the yeast S. cerevisiae with microtubule- destabilizing drugs, which led to the identification of Bub1 as a mitotic checkpoint protein [Roberts BT et al., Mol. Cell Biol., 14, 8282, 1994]. A number of recent publications provide evidence that Bub1 plays multiple roles during mitosis which, have been reviewed by Elowe [Elowe S, Mol. Cell. Biol. 31 , 3085, 201 1 ]. In particular, Bub1 is one of the first mitotic checkpoint proteins that binds to the kinetochores of duplicated chromosomes and probably acts as a scaffolding protein to constitute the mitotic checkpoint complex. Furthermore, via phosphorylation of histone H2A, Bub1 localizes the protein shugoshin to the centromeric region of the chromosomes to prevent premature segregation of the paired chromosomes [Kawashima et al. Science 327, 172, 2010]. In addition, together with a Thr-3 phosphorylated Histone H3 the shugoshin protein functions as a binding site for the chromosomal passenger complex which includes the proteins survivin, borealin, INCENP and Aurora B. The chromosomal passenger complex is seen as a tension sensor within the mitotic checkpoint mechanism, which dissolves erroneously formed microtubule-kinetochor attachments such as syntelic (both sister kinetochors are attached to one spindle pole) or merotelic (one kinetochor is attached to two spindle poles) attachments [Watanabe Y, Cold Spring Harb. Symp. Quant. Biol. 75, 419, 2010]. Recent data suggest that the phosphorylation of histone H2A at Thr 121 by Bub1 kinase is sufficient to localize AuroraB kinase to fulfill the attachment error correction checkpoint [Ricke et al. J. Cell Biol. 199, 931 -949, 2012].
Incomplete mitotic checkpoint function has been linked with aneuploidy and tumourigenesis [Weaver BA and Cleveland DW, Cancer Res. 67, 10103, 2007; King RW, Biochim Biophys Acta 1786, 4, 2008]. In contrast, complete inhibition of the mitotic checkpoint has been recognised to result in severe chromosome missegregation and induction of apoptosis in tumour cells [Kops GJ et al., Nature Rev. Cancer 5, 773, 2005; Schmidt M and Medema RH, Cell Cycle 5, 159, 2006; Schmidt M and Bastians H, Drug Res. Updates 10, 162, 2007]. Thus, mitotic checkpoint abrogation through pharmacological inhibition of components of the mitotic checkpoint, such as Bub1 kinase, represents a new approach for the treatment of proliferative disorders, including solid tumours such as carcinomas, sarcomas, leukaemias and lymphoid malignancies or other disorders, associated with uncontrolled cellular proliferation.
The present invention relates to chemical compounds that inhibit Bub1 kinase.
Established anti-mitotic drugs such as vinca alkaloids, taxanes or epothilones activate the mitotic checkpoint, inducing a mitotic arrest either by stabilising or destabilising microtubule dynamics. This arrest prevents separation of the duplicated chromosomes to form the two daughter cells. Prolonged arrest in mitosis forces a cell either into mitotic exit without cytokinesis (mitotic slippage or adaption) or into mitotic catastrophe leading to cell death [Rieder CL and Maiato H, Dev. Cell 7, 637, 2004]. In contrast, inhibitors of Bub1 prevent the establishment and/or functionality of the mitotic checkpoint and/or microtubule-kinetochor attachment error correction mechanisms, which finally results in severe chromosomal missegregation, induction of apoptosis and cell death [Baron A et al. eLife 2016;5:e12187]. These findings suggest that Bub1 inhibitors should be of therapeutic value for the treatment of proliferative disorders associated with enhanced uncontrolled proliferative cellular processes such as, for example, cancer, inflammation, arthritis, viral diseases, cardiovascular diseases, or fungal diseases in a warm-blooded animal such as man.
WO 2013/050438, WO 2013/092512, WO 2013/167698 disclose substituted benzylindazoles, substituted benzylpyrazoles and substituted benzylcycloalkyl- pyrazoles, respectively, which are Bub1 kinase inhibitors.
WO 2014/147203, WO 2014/147204, WO2014202590, WO2014202588, WO2014202584, WO2014202583, and WO2015/063003, disclose substituted indazoles, substituted pyrazoles, and substituted cycloalkylpyrazoles, which are Bub1 kinase inhibitors.
Due to the fact that especially cancer disease as being expressed by uncontrolled proliferative cellular processes in tissues of different organs of the human- or animal body still is not considered to be a controlled disease in that sufficient drug therapies already exist, there is a strong need to provide further new therapeutically useful drugs, preferably inhibiting new targets and providing new therapeutic options (e.g. drugs with improved pharmacological properties, such as improved target Bub1 inhibition potency).
Description of the invention
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 Bub1 .
Inhibitors of Bub1 represent valuable compounds that should complement therapeutic options either as single agents or in combination with other drugs.
In accordance with a first aspect, the invention covers compounds of general formula (I),
Figure imgf000007_0001
in which
V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH or CR5, or,
V and Y represent N, and W and Z, independently of each other, represent CH or CR5,
R1 and R2 represent, independently of each other, a hydrogen atom or a halogen atom,
R3 represents a hydrogen atom or a group selected from:
hydroxy, Ci-C6-alkyl, Ci-C6-haloalkyl, Ci-C6-hydroxyalkyl,
(Ci-C3-alkoxy)-(Ci-C6-alkyl)-, Ca-Ce-cycloalkyl, (Ca-Ce-cycloalkylMCi-Ca-alkyl)-, Ci-Ce-alkoxy, Ci-C6-haloalkoxy, (C2-C6-hydroxyalkyl)-0-, (Ci-C3-alkoxy)-(C2-C6- alkoxy)-, C3-C6-cycloalkoxy and (C3-C6-cycloalkyl)-(Ci-C3-alkoxy)-,
wherein said hydroxyalkyl groups are optionally substituted with one, two or three halogen atoms selected from:
fluorine and chlorine, represents a hydrogen atom, or a group selected from:
Ci-Ce-alkyl, C3-C6-cycloalkyl, Ci-C6-haloalkyl, C2-C6-hydroxyalkyl and
(Ci-C3-alkoxy)-(C2-C6-alkyl)-, R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci-C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy,
(C2-C3-hydroxyalkyl)-0-, (Ci-C3-alkoxy)-(C2-C3-alkoxy)-, Ci-C3-haloalkoxy, -N(H)C(=0)H,
Figure imgf000008_0001
-N(H)C(=0)-(C3-C4-cycloalkyl),
-N(H)C(=0)-(Ci-C3-hydroxyalkyl), -C(=0)N(R9)2, -N(H)C(=0)N(H)R9 and -C(=0)OR10,
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from:
*-X-(CH2)2-#,
*-CH2-X-CH2-#,
*-(CH2)2-X-#,
*-X-(CH2)3-#,
*-CH2-X-(CH2)2-#,
*-(CH2)2-X-CH2-#,
*-(CH2)3-X-#,
*-C(R11)=N-C(R11)=C(R11)-#,
and
*-C(R11)=C(R11)-C(R11)=N-#,
in which groups X represents -NH-, -N(Ci-C3-alkyl)-, -0-, -S-, -S(=0)- or
-S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule, represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl, Ci-C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -C3-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-,
R10 represents a hydrogen atom or a group selected from:
Ci-C3-alkyl, Ci-C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -C3-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-, R1 1 represents, independently of each other, a hydrogen atom or a halogen atom or a group selected from:
methyl, ethyl and Ci -C2-haloalkyl, or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
A second aspect of the invention are compounds of formula (I) as defined herein, wherein
V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH or CR5,
R1 and R2 represent, independently of each other, a hydrogen atom or a halogen atom,
R3 represents a hydrogen atom or a group selected from:
Ci -Ce-alkoxy, Ci -C6-haloalkoxy, (C2-C6-hydroxyalkyl)-0-, (Ci -C3-alkoxy)-(C2-C6- alkoxy)-, C3-C6-cycloalkoxy and (C3-C6-cycloalkyl)-(Ci -C3-alkoxy)-, R4 represents a hydrogen atom, or a group selected from:
Ci -Ce-alkyl, C3-C6-cycloalkyl, Ci -C6-haloalkyl, C2-C6-hydroxyalkyl and (Ci -C3-alkoxy)-(C2-C6-alkyl)-,
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci -C3-alkyl, C3-C4-cycloalkyl, Ci -C3-haloalkyl, Ci -C3-alkoxy, C1 -C3- haloalkoxy and -C(=0)N(R9)2,
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from:
*-X-(CH2)3-#,
Figure imgf000010_0001
*-(CH2)3-X-#,
*-C(R11)=N-C(R11)=C(R11)-#,
and
*-C(R11)=C(R11)-C(R11)=N-#,
in which groups X represents -NH-, -N(Ci-C3-alkyl)-, -0-, -S-, -S(=0)- or
-S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule,
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl and (Ci-C3-alkoxy)-(C2-C3- alkyl)-,
R11 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group, or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
A third aspect of the invention are compounds of formula (I) as defined herein, wherein
V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH or CR5, R1 and R2 represent, independently of each other, a hydrogen atom or a halogen atom, R3 represents a hydrogen atom or a group selected from:
Ci-C3-alkoxy, (C2-C3-hydroxyalkyl)-0- and (Ci-C3-alkoxy)-(C2-C3-alkoxy)-,
R4 represents a hydrogen atom, or a group selected from:
Ci-C3-alkyl and C2-C3-hydroxyalkyl,
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci-C3-alkyl, Ci-C3-alkoxy and -C(=0)N(R9)2,
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from:
Figure imgf000011_0001
*-C(R11)=N-C(R11)=C(R11)-#,
and
*-C(R11)=C(R11)-C(R11)=N-#,
in which groups X represents -0-, -S-, or -S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule,
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl and C3-C4-cycloalkyl,
R11 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group, or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
A fourth aspect of the invention are compounds of formula (I) as defined herein, wherein V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH or CR5,
R1 and R2 represent, independently of each other, a halogen atom,
R3 represents a group selected from:
Ci-C3-alkoxy, (C2-C3-hydroxyalkyl)-0- and (Ci-C3-alkoxy)-(C2-C3-alkoxy)-,
R4 represents a
Ci-C3-alkyl group,
R5 represents, independently of each other, a group selected from:
Ci-C3-alkyl and Ci-C3-alkoxy,
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from:
Figure imgf000012_0001
*-CH=N-CH=CH-#,
and
*-CH=CH-CH=N-#,
in which groups X represents -0-, -S-, or -S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule, or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
A fifth aspect of the invention are compounds of formula (I) as defined herein, wherein V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH,
R1 and R2 represent a fluorine atom,
R3 represents a group selected from:
ethoxy, 2-hydroxyethoxy and 2-methoxyethoxy,
R4 represents a methyl group,
R5 represents, independently of each other, a group selected from:
methyl and methoxy,
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from:
Figure imgf000013_0001
*-CH=N-CH=CH-#,
and
*-CH=CH-CH=N-#,
in which groups X represents -0-, -S-, or -S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of Rto the rest of the molecule, 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 a further aspect of the invention compounds of formula (I) as described above are selected from the group consisting of:
2-{1 -[2,6-difluoro-4-(2-methoxyethoxy)benzyl]-1 H-pyrazolo[4,3-b]pyridin-3-yl}-5- methoxy-N-(3-methoxypyridin-4-yl)pyrimidin-4-amine, 2-[1 -(4-ethoxy-2,6-difluorobenzyl)-1 H-pyrazolo[3,4-c]pyridin-3-yl]-5-methoxy-N-(3- methoxypyridin-4-yl)pyrimidin-4-amine,
2-[1 -(4-ethoxy-2,6-difluorobenzyl)-1 H-pyrazolo[3,4-c]pyridin-3-yl]-5-methoxy-N-(2- methylpyrimidin-4-yl)pyrimidin-4-amine,
N-(2-{1 -[2,6-difluoro-4-(2-methoxyethoxy)benzyl]-1 H-pyrazolo[4,3-b]pyridin-3-yl}-5- methoxypyrimidin-4-yl)pyridazin-4-amine,
2-[1 -(4-ethoxy-2,6-difluorobenzyl)-1 ,4,6,7-tetrahydrothiopyrano[4,3-c]pyrazol-3-yl]-5- methoxy-N-(pyridin-4-yl)pyrimidin-4-amine,
2-[1 -(4-ethoxy-2,6-difluorobenzyl)-1 ,4,6,7-tetrahydropyrano[4,3-c]pyrazol-3-yl]-5- methoxy-N-(pyridin-4-yl)pyrimidin-4-amine,
2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl)amino]pyrimidin-2-yl}-^ H- pyrazolo[4,3-b]pyridin-1 -yl)methyl]phenoxy}ethanol,
2-[3,5-difluoro-4-({3-[5-methoxy-4-(pyridazin-4-ylamino)pyrimidin-2-yl]-1 H-pyrazolo[4,3- b]pyridin-1 -yl}methyl)phenoxy]ethanol, and
2-[1 -(4-ethoxy-2,6-difluorobenzyl)-5,5-dioxido-1 ,4,6,7-tetrahydrothiopyrano[4,3-c]- pyrazol-3-yl]-5-methoxy-N-(pyridin-4-yl)pyrimidin-4-amine,
or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
A further aspect of the invention are compounds of formula (I), wherein
V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH or CR5, or,
V and Y represent N, and W and Z, independently of each other, represent CH or CR5 and
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci -C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy,
(C2-C3-hydroxyalkyl)-0-, (Ci -C3-alkoxy)-(C2-C3-alkoxy)-, Ci-C3-haloalkoxy, -N(H)C(=0)H,
Figure imgf000014_0001
-N(H)C(=0)-(C3-C4-cycloalkyl), -N(H)C(=0)-(Ci-C3-hydroxyalkyl), -C(=0)N(R9)2, -N(H)C(=0)N(H)R9 and -C(=0)OR10, and
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl, Ci-C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -C3-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-, and
R10 represents a hydrogen atom or a group selected from:
Ci-C3-alkyl, Ci-C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -C3-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-.
Yet another aspect of the invention are compounds of formula (I), in which
V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH or CR5, and
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci-C3-alkyl, C3-C -cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy, Ci-C3- haloalkoxy and -C(=0)N(R9)2,
and
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl, C2-C3-hydroxyalkyl, C3-C -cycloalkyl and (Ci-C3-alkoxy)-(C2-C3- alkyl)-.
Yet another aspect of the invention are compounds of formula (I), in which
V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH or CR5, and R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci-C3-alkyl, Ci-C3-alkoxy and -C(=0)N(R9)2,
and
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl and C3-C4-cycloalkyl.
Yet another aspect of the invention are compounds of formula (I), in which
V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH or CR5, and
R5 represents, independently of each other, a group selected from:
Ci-C3-alkyl and Ci-C3-alkoxy.
Yet another aspect of the invention are compounds of formula (I), in which
V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH or CR5, and
R5 represents, independently of each other, a group selected from:
methyl and methoxy.
A further aspect of the invention are compounds of formula (I), wherein
V, W, Y and Z, independently of each other, represent CH or CR5.
Yet another aspect of the invention are compounds of formula (I), in which:
one of V, W, Y and Z represents CR5, and the others represent CH.
Yet another aspect of the invention are compounds of formula (I), in which:
V represents N, and one of W, Y and Z, represents CR5, and the others represent CH.
A further aspect of the invention are compounds of formula (I), wherein
V, W, Y and Z, independently of each other, represent CH or CR5,
and
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci-C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy,
(C2-C3-hydroxyalkyl)-0-, (Ci-C3-alkoxy)-(C2-C3-alkoxy)-, Ci-C3-haloalkoxy, -N(H)C(=0)H,
Figure imgf000017_0001
-N(H)C(=0)-(C3-C4-cycloalkyl), -N(H)C(=0)-(Ci-C3-hydroxyalkyl), -C(=0)N(R9)2, -N(H)C(=0)N(H)R9 and -C(=0)OR10,
and
R9 represents, independently of each other, a hydrogen atom or a group selected from :
Ci-C3-alkyl, Ci-C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -Cs-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-, and
R10 represents a hydrogen atom or a group selected from :
Ci-C3-alkyl, Ci-C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C -cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -C3-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-.
Yet another aspect of the invention are compounds of formula (I), in which
V, W, Y and Z, independently of each other, represent CH or CR5,
and
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci-C3-alkyl, C3-C -cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy, C1 -C3- haloalkoxy and -C(=0)N(R9)2,
and
R9 represents, independently of each other, a hydrogen atom or a group selected from :
Ci-C3-alkyl, C2-C3-hydroxyalkyl, C3-C -cycloalkyl and (Ci-C3-alkoxy)-(C2-C3- alkyl)-.
Yet another aspect of the invention are compounds of formula (I), in which V, W, Y and Z, independently of each other, represent CH or CR5,
and
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci-C3-alkyl, Ci-C3-alkoxy and -C(=0)N(R9)2,
and
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl and C3-C4-cycloalkyl.
Yet another aspect of the invention are compounds of formula (I), in which
V, W, Y and Z, independently of each other, represent CH or CR5,
and
R5 represents, independently of each other, a group selected from:
Ci-C3-alkyl and Ci-C3-alkoxy.
Yet another aspect of the invention are compounds of formula (I), in which
V, W, Y and Z, independently of each other, represent CH or CR5,
and
R5 represents a methoxy group.
A further aspect of the invention are compounds of formula (I), wherein
V represents N, and W, Y and Z, independently of each other, represent CH or CR5.
A further aspect of the invention are compounds of formula (I), wherein
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, and
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci-C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy
(C2-C3-hydroxyalkyl)-0-, (Ci-C3-alkoxy)-(C2-C3-alkoxy)-, Ci-C3-haloalkoxy, -N(H)C(=0)H,
Figure imgf000018_0001
-N(H)C(=0)-(C3-C4-cycloalkyl), -N(H)C(=0)-(Ci-C3-hydroxyalkyl), -C(=0)N(R9)2, -N(H)C(=0)N(H)R9 and -C(=0)OR10,
and R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl, Ci -C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -C3-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-, and
R10 represents a hydrogen atom or a group selected from:
Ci-C3-alkyl, Ci -C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -C3-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-.
Yet another aspect of the invention are compounds of formula (I), in which
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, and
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci -C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy, C1 -C3- haloalkoxy and -C(=0)N(R9)2,
and
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl and (Ci-C3-alkoxy)-(C2-C3- alkyl)-.
Yet another aspect of the invention are compounds of formula (I), in which
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, and
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci -C3-alkyl, Ci-C3-alkoxy and -C(=0)N(R9)2,
and
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl and C3-C4-cycloalkyl.
Yet another aspect of the invention are compounds of formula (I), in which
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, R5 represents, independently of each other, a group selected from:
Ci-C3-alkyl and Ci-C3-alkoxy.
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, and
R5 represents a methyl group.
A further aspect of the invention are compounds of formula (I), wherein
W represents N, and V, Y and Z, independently of each other, represent CH or CR5.
A further aspect of the invention are compounds of formula (I), wherein
W represents N, and V, Y and Z, independently of each other, represent CH or CR5, and
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci-C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy,
(C2-C3-hydroxyalkyl)-0-, (Ci-C3-alkoxy)-(C2-C3-alkoxy)-, Ci-C3-haloalkoxy, -N(H)C(=0)H,
Figure imgf000020_0001
-N(H)C(=0)-(C3-C4-cycloalkyl), -N(H)C(=0)-(Ci-C3-hydroxyalkyl), -C(=0)N(R9)2, -N(H)C(=0)N(H)R9 and -C(=0)OR10,
and
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl, Ci-C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -C3-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-, and
R10 represents a hydrogen atom or a group selected from:
Ci-C3-alkyl, Ci-C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C -cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -C3-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-.
Yet another aspect of the invention are compounds of formula (I), in which
W represents N, and V, Y and Z, independently of each other, represent CH or CR5, and
R5 represents, independently of each other, a halogen atom or a group selected from: cyano, Ci -C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy, C1 -C3- haloalkoxy and -C(=0)N(R9)2,
and
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl and (Ci-C3-alkoxy)-(C2-C3- alkyl)-.
Yet another aspect of the invention are compounds of formula (I), in which
W represents N, and V, Y and Z, independently of each other, represent CH or CR5, and
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci -C3-alkyl, Ci-C3-alkoxy and -C(=0)N(R9)2,
and
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl and C3-C4-cycloalkyl.
Yet another aspect of the invention are compounds of formula (I), in which
W represents N, and V, Y and Z, independently of each other, represent CH or CR5, and
R5 represents, independently of each other, a group selected from:
Ci -C3-alkyl and Ci-C3-alkoxy.
Yet another aspect of the invention are compounds of formula (I), in which
W represents N, and V, Y and Z represent CH.
Yet another aspect of the invention are compounds of formula (I), in which
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci -C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy,
(C2-C3-hydroxyalkyl)-0-, (Ci -C3-alkoxy)-(C2-C3-alkoxy)-, Ci-C3-haloalkoxy, -N(H)C(=0)H,
Figure imgf000021_0001
-N(H)C(=0)-(C3-C4-cycloalkyl), -N(H)C(=0)-(Ci-C3-hydroxyalkyl), -C(=0)N(R9)2, -N(H)C(=0)N(H)R9 and -C(=0)OR10. Yet another aspect of the invention are compounds of formula (I), in which
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci-C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy, C1 -C3- haloalkoxy and -C(=0)N(R9)2.
Yet another aspect of the invention are compounds of formula (I), in which
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci-C3-alkyl, Ci-C3-alkoxy and -C(=0)N(R9)2.
Yet another aspect of the invention are compounds of formula (I), in which
R5 represents, independently of each other, a group selected from :
Ci-C3-alkyl and Ci-C3-alkoxy.
Yet another aspect of the invention are compounds of formula (I), in which
R5 represents, independently of each other, a group selected from :
methyl and methoxy.
Yet another aspect of the invention are compounds of formula (I), in which
R9 represents, independently of each other, a hydrogen atom or a group selected from :
Ci-C3-alkyl, Ci-C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -C3-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-.
Yet another aspect of the invention are compounds of formula (I), in which
R9 represents, independently of each other, a hydrogen atom or a group selected from :
Ci-C3-alkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl and (Ci-C3-alkoxy)-(C2-C3- alkyl)-.
Yet another aspect of the invention are compounds of formula (I), in which
R9 represents, independently of each other, a hydrogen atom or a group selected from :
Ci-C3-alkyl and C3-C -cycloalkyl. Yet another aspect of the invention are compounds of formula (I), in which
R9 represents, independently of each other, a hydrogen atom or a methyl group. Yet another aspect of the invention are compounds of formula (I), in which
R10 represents a hydrogen atom or a group selected from:
Ci -C3-alkyl, Ci -C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -C3-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-. Yet another aspect of the invention are compounds of formula (I), in which
R1 and R2 represent, independently of each other, a halogen atom, preferably a fluorine atom.
Yet another aspect of the invention are compounds of formula (I), in which
R1 and R2 represent, independently of each other, a hydrogen atom or a halogen atom,
and
R3 represents a hydrogen atom or a group selected from:
hydroxy, Ci -Ce-alkyl, Ci -Ce-haloalkyl, Ci -Ce-hydroxyalkyl,
(Ci -C3-alkoxy)-(Ci -C6-alkyl)-, C3-C6-cycloalkyl, (C3-C6-cycloalkyl)-(Ci -C3-alkyl)-,
Ci -Ce-alkoxy, Ci -C6-haloalkoxy, (C2-C6-hydroxyalkyl)-0-, (Ci -C3-alkoxy)-(C2-C6- alkoxy)-, C3-C6-cycloalkoxy and (C3-C6-cycloalkyl)-(Ci -C3-alkoxy)-,
wherein said hydroxyalkyl groups are optionally substituted with one, two or three halogen atoms selected from:
fluorine and chlorine.,
Yet another aspect of the invention are compounds of formula (I), in which
R1 and R2 represent, independently of each other, a hydrogen atom or a halogen atom,
and
R3 represents a hydrogen atom or a group selected from:
Ci -Ce-alkoxy, Ci -Ce-haloalkoxy, (C2-C6-hydroxyalkyl)-0-, (Ci -C3-alkoxy)-(C2-C6- alkoxy)-, C3-C6-cycloalkoxy and (C3-C6-cycloalkyl)-(Ci -C3-alkoxy)-. Yet another aspect of the invention are compounds of formula (I), in which R1 and R2 represent, independently of each other, a hydrogen atom or a halogen atom,
and
R3 represents a hydrogen atom or a group selected from:
Ci -C3-alkoxy, (C2-C3-hydroxyalkyl)-0- and (Ci-C3-alkoxy)-(C2-C3-alkoxy)-.
Yet another aspect of the invention are compounds of formula (I), in which
R1 and R2 represent, independently of each other, a halogen atom,
and
R3 represents a group selected from:
Ci -C3-alkoxy, (C2-C3-hydroxyalkyl)-0- and (Ci-C3-alkoxy)-(C2-C3-alkoxy)-.
Yet another aspect of the invention are compounds of formula (I), in which
R1 and R2 represent a fluorine atom,
and
R3 represents a group selected from:
ethoxy, 2-hydroxyethoxy and 2-methoxyethoxy.
Yet another aspect of the invention are compounds of formula (I), in which
R4 represents a hydrogen atom, or a group selected from:
Ci-Ce-alkyl, C3-C6-cycloalkyl, Ci-C6-haloalkyl, C2-C6-hydroxyalkyl and
(Ci-C3-alkoxy)-(C2-C6-alkyl)-.
Yet another aspect of the invention are compounds of formula (I), in which
R4 represents a hydrogen atom, or a group selected from:
Ci-C3-alkyl and C2-C3-hydroxyalkyl.
Yet another aspect of the invention are compounds of formula (I), in which represents a Ci-C3-alkyl group.
Yet another aspect of the invention are compounds of formula (I), in which
R4 represents a methyl group. Yet another aspect of the invention are compounds of formula (I), in which R7 and R8 are linked to one another in such a way that they jointly form a group selected from:
*-X-(CH2)2-#,
Figure imgf000025_0001
*-(CH2)2-X-#,
*-X-(CH2)3-#,
*-CH2-X-(CH2)2-#,
*-(CH2)2-X-CH2-#,
*-(CH2)3-X-#,
*-C(R11)=N-C(R11)=C(R11)-#,
and
*-C(R11)=C(R11)-C(R11)=N-#,
in which groups X represents -NH-, -N(Ci-C3-alkyl)-, -0-, -S-, -S(=0)- or
-S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule,
and
R11 represents, independently of each other, a hydrogen atom or a halogen atom or a group selected from:
methyl, ethyl and Ci-C2-haloalkyl.
Yet another aspect of the invention are compounds of formula (I), in which
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from:
*-X-(CH2)3-#,
*-CH2-X-(CH2)2-#,
*-(CH2)2-X-CH2-#,
*-(CH2)3-X-#,
*-C(R11)=N-C(R11)=C(R11)-#,
and
*-C(R11)=C(R11)-C(R11)=N-#,
in which groups X represents -NH-, -N(Ci-C3-alkyl)-, -0-, -S-, -S(=0)- or
-S(=0)2-,
and, in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule,
and
R1 1 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group.
Yet another aspect of the invention are compounds of formula (I), in which
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from :
Figure imgf000026_0001
*-C(R1 1 )=N-C(R1 1)=C(R1 1)-#,
and
*-C(R1 1 )=C(R1 1 )-C(R1 1)=N-#,
in which groups X represents -0-, -S-, or -S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule,
and
R1 1 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group.
Yet another aspect of the invention are compounds of formula (I), in which
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from :
Figure imgf000026_0002
*-CH=N-CH=CH-#,
and
*-CH=CH-CH=N-#,
in which groups X represents -0-, -S-, or -S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule, Yet another aspect of the invention are compounds of formula (I), in which
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from :
*-X-(CH2)2-#,
Figure imgf000027_0001
*-(CH2)2-X-#,
*-X-(CH2)3-#,
Figure imgf000027_0002
and
*-(CH2)3-X-#,
in which groups X represents -NH-, -N(Ci-C3-alkyl)-, -0-, -S-, -S(=0)- or
-S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule,
Yet another aspect of the invention are compounds of formula (I), in which
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from :
*-X-(CH2)3-#,
Figure imgf000027_0003
and
*-(CH2)3-X-#,
in which groups X represents -NH-, -N(Ci-C3-alkyl)-, -0-, -S-, -S(=0)- or
-S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule.
Yet another aspect of the invention are compounds of formula (I), in which R7 and R8 are linked to one another in such a way that they jointly form a group selected from :
Figure imgf000028_0001
in which group X represents -0-, -S-, or -S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule.
Yet another aspect of the invention are compounds of formula (I), in which
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from :
*-C(R1 1 )=N-C(R1 1)=C(R1 1)-#,
and
*-C(R1 1 )=C(R1 1 )-C(R1 1)=N-#,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule,
and
R1 1 represents, independently of each other, a hydrogen atom or a halogen atom or a group selected from :
methyl, ethyl and Ci-C2-haloalkyl.
Yet another aspect of the invention are compounds of formula (I), in which
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from :
*-C(R1 1 )=N-C(R1 1)=C(R1 1)-#,
and
*-C(R1 1 )=C(R1 1 )-C(R1 1)=N-#,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule,
and
R1 1 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group. Yet another aspect of the invention are compounds of formula (I), in which
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from:
*-CH N-CH=CH-#,
and
*-CH CH-CH=N-#,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule.
Yet another aspect of the invention are compounds of formula (I), in which
R11 represents, independently of each other, a hydrogen atom or a halogen atom or a group selected from:
methyl, ethyl and Ci-C2-haloalkyl.
Yet another aspect of the invention are compounds of formula (I), in which
R11 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group.
Yet another aspect of the invention are compounds of formula (I), in which
R11 represents a hydrogen atom.
One aspect of the invention are compounds of formula (I) as described in the examples, as characterized by their names in the title, as claimed in claim 6, and their structures as well as the subcombinations of all residues specifically disclosed in the compounds of the examples.
Another aspect of the present invention are the intermediates as used for their synthesis.
A further aspect of the invention are compounds of formula (I), which are present as their salts.
Yet another aspect of the invention are compounds of formula (I) in which,
the salt is a pharmaceutically acceptable salt. 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.
Another embodiment of the invention are compounds according to the claims as disclosed in the Claims section wherein the definitions are limited according to the preferred or more preferred definitions as disclosed below or specifically disclosed residues of the exemplified compounds and subcombinations thereof.
In accordance with a further aspect, the present invention relates to intermediate compounds which are useful in the preparation of compounds of the present invention of general formula (I), particularly in the method described herein.
In particular, the present invention relates to a compound of general formula (II) :
Figure imgf000030_0001
in which R1 , R2, R3, R4, R7 and R8 are as defined herein. ln accordance with yet another aspect, the present invention relates to the use of an intermediate compound of general formula (II) :
Figure imgf000031_0001
in which R1 , R2, R3, R4, R7 and R8 are as defined herein, for the preparation of a compound of general formula (I) as defined supra.
In accordance with yet another aspect, the present invention relates to the use of an intermediate compound of general formula (III) :
V - N
in which V, W, Y and Z are as defined herein, and X2 represents F, CI, Br, I, boronic acid or a boronic acid ester, such as for example 4,4,5,5-tetramethyl-2-phenyl-1 ,3,2- dioxaborolane (boronic acid pinacole ester), for the preparation of a compound of general formula (I) as defined supra.
In accordance with yet another aspect, the present invention relates to a method of preparing a compound of general formula (I) as defined supra, said method comprising the step of allowing an intermediate compound of general formula (II) :
Figure imgf000032_0001
in which R1 , R2, R3, R4, R7 and R8 are as defined herein, to react with a compound of general formula (III) :
Figure imgf000032_0002
in which V, W, Y and Z are as defined herein, and X2 represents F, CI, Br, I, boronic acid or a boronic acid ester, such as for example 4,4,5,5-tetramethyl-2-phenyl-1 ,3,2- dioxaborolane (boronic acid pinacole ester),
thereby giving a compound of general formula (I) :
Figure imgf000032_0003
in which in which R\ R2, R3, R4, R7, R8, V, W, Y ant Z are as defined herein. In accordance with yet another aspect, the present invention relates to a method of preparing a compound of general formula (l-b) as defined herein, said method comprising the step of allowing a compound of general formula (l-a) :
Figure imgf000033_0001
(l-a) , in which R1 , R2, R4, R7, R8, V, W, Y and Z are as defined herein, and R12 represents a methyl group to react with a reagent such as for example boron tribromide,
thereby giving a compound of general formula (l-b) :
Figure imgf000033_0002
(l-b) , in which R1 , R2, R4, R7, R8, V, W, Y and Z are as defined herein. 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. For example, whenever R1 , R2, R4, R5, R6, R7, R8, R9, R10, R1 1 , V, W, Y and/or Z occur more than one time for any compound of formula (I) each definition of R1 , R2, R3, R4, R5, R6, R7, R8, R9, R10, R1 1 , V, W, Y and/or Z is independent.
As used herein, an oxo substituent represents an oxygen atom, which is bound to a carbon atom or to a sulfur atom via a double bond.
Should a composite substituent be composed of more than one parts, e.g. (C1 -C3- alkoxy)-(Ci-C6-alkyl), (Cs-Ce-cycloalkylHCi-Cs-alkyl)- or (Cs-Ce-cycloalkylHCi-Cs- alkoxy)-, it is possible for the position of a given part to be at any suitable position of said composite substituent, i.e. the Cs-Ce-cycloalkyl part can be attached to any carbon atom of the Ci-C3-alkyl part of said (C3-C6-cycloalkyl)-(Ci -C3-alkyl)- group. A hyphen at the beginning or at the end of such a composite substituent indicates the point of attachment of said composite substituent to the rest of the molecule. Should a ring, comprising carbon atoms and optionally one or more heteroatoms, such as nitrogen, oxygen or sulfur atoms for example, be substituted with a substituent, it is possible for said substituent to be bound at any suitable position of said ring, be it bound to a suitable carbon atom and/or to a suitable heteroatom.
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.
"suitable" within the sense of the invention means chemically possible to be made by methods within the knowledge of a skilled person.
The terms as mentioned in the present text have preferably the following meanings :
The term "halogen atom", "halo-" or "Hal-" is to be understood as meaning a fluorine, chlorine, bromine or iodine atom. The term "Ci -C6-alkyl" is to be understood as meaning a linear or branched, saturated, monovalent hydrocarbon group having 1 , 2, 3, 4, 5 or 6 carbon atoms, e.g. a methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, iso-propyl, iso-butyl, sec-butyl, tert-butyl, iso-pentyl, 2-methylbutyl, 1 -methylbutyl, 1 -ethylpropyl, 1 ,2-dimethylpropyl, neo-pentyl, 1 ,1 -dimethylpropyl, 4-methylpentyl, 3-methylpentyl, 2-methylpentyl, 1 -methylpentyl, 2- ethylbutyl, 1 -ethylbutyl, 3,3-dimethylbutyl, 2,2-dimethylbutyl, 1 ,1 -dimethylbutyl, 2,3- dimethylbutyl, 1 ,3-dimethylbutyl, or 1 ,2-dimethylbutyl group, or an isomer thereof. Particularly, said group has 1 , 2, 3, 4 or 5 carbon atoms ("Ci-C5-alkyl"), e.g. a methyl, ethyl, propyl, butyl, iso-propyl, iso-butyl, sec-butyl, tert-butyl, iso-pentyl, 2-methylbutyl, 1 -methylbutyl, 1 -ethylpropyl, 1 ,2-dimethylpropyl, neo-pentyl, 1 ,1 -dimethylpropyl group, more particularly 1 , 2 or 3 carbon atoms ("Ci-C3-alkyl"), e.g. a methyl, ethyl, n-propyl- or iso-propyl group.
The term "Ci-C6-haloalkyl" is to be understood as meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "Ci-C6-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 Ci-Ce-haloalkyl group is, for example, -CF3, - CHF2, -CH2F, -CF2CF3, -CH2CH2F, -CH2CHF2, -CH2CF3, -CH2CH2CF3, or -CH(CH2F)2. Particularly, said group has 1 , 2, 3 or 4 carbon atoms ("Ci-C4-haloalkyl"), more particularly 1 , 2 or 3 carbon atoms ("Ci-C3-haloalkyl"), even more particularly 1 or 2 carbon atoms ("Ci-C2-haloalkyl").
The term "Ci -C6-alkoxy" is to be understood as meaning a linear or branched, saturated, monovalent, hydrocarbon group of formula -O-alkyl, in which the term "alkyl" is defined supra, e.g. a methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, iso-butoxy, tert-butoxy, sec-butoxy, pentoxy, iso-pentoxy, or n-hexoxy group, or an isomer thereof. Particularly, said group has 1 , 2, 3 or 4 carbon atoms ("Ci-C4-alkoxy"), more particularly 1 , 2 or 3 carbon atoms ("Ci-C3-alkoxy").
The term "Ci-Ce-haloalkoxy" is to be understood as meaning a linear or branched, saturated, monovalent Ci -C6-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 Ci-C6-haloalkoxy group is, for example, - OCF3, -OCHF2, -OCH2F, -OCF2CF3, or -OCH2CF3. Particularly, said group has 1 , 2, 3 or 4 carbon atoms ("Ci-C4-haloalkoxy"), more particularly 1 , 2 or 3 carbon atoms ("C1-C3- haloalkoxy").
The term "Ci-Ce-hydroxyalkyI" is to be understood as preferably meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "Ci-C6-alkyl" 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, 3- hydroxy-2-methyl-propyl, 2-hydroxy-2-methyl-propyl, 1 -hydroxy-2-methyl-propyl group. Particularly, said group has 1 , 2, 3 or 4 carbon atoms ("Ci-C4-hydroxyalkyl"), more particularly 1 , 2 or 3 carbon atoms ("Ci-C3-hydroxyalkyl").
The term "Cs-Ce-cycloalkyl" is to be understood as meaning a saturated, monovalent, monocyclic hydrocarbon ring which contains 3, 4, 5 or 6 carbon atoms ("C3-C6- cycloalkyl"). Said C3-C6-cycloalkyl group is for example, a monocyclic hydrocarbon ring, e.g. a cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl ring. Particularly, said group has 3 or 4 carbon atoms ("C3-C4-cycloalkyl").
The term "Cs-Ce-cycloalkoxy" means a saturated, monovalent, monocyclic group of formula (C3-C6-cycloalkyl)-0-, which contains 3, 4, 5 or 6 carbon atoms, in which the term "Cs-Ce-cycloalkyl" is defined supra, e.g. a cyclopropyloxy, cyclobutyloxy, cyclopentyloxy or cyclohexyloxy group.
The term "Ci-C6", as used in the present text, e.g. in the context of the definition of "Ci-Ce-alkyl", "Ci-C6-haloalkyl", "Ci-Ce-hydroxyalkyI", "Ci-C6-alkoxy" or "Ci-C6- haloalkoxy" means a group having a finite number of carbon atoms of 1 to 6, i.e. 1 , 2, 3, 4, 5 or 6 carbon atoms.
Further, as used herein, the term "C3-C6", as used in the present text, e.g. in the context of the definition of "C3-C6-cycloalkyl", means a cycloalkyl group having a finite number of carbon atoms of 3 to 6, i.e. 3, 4, 5 or 6 carbon atoms.
When a range of values is given, said range encompasses each value and sub-range within said range.
For example: "d-Ce" encompasses Ci , C2, C3, C4, C5, Ce, C1 -C6, C1 -C5, C1 -C4, C1 -C3, C1 -C2, C2-C6, C2-C5, C2-C4, C2-C3, C3-C6, C3-C5, C3-C4, C4-C6, C4-C5, and C5-C6;
"C1 -C3" encompasses Ci , C2, C3, C1 -C3, C1 -C2 and C2-C3;
"C2-C6" encompasses C2, C3, C4, C5, Ce, C2-C6, C2-C5, C2-C4, C2-C3, C3-C6, C3-C5, C3-C4, C4-C6, C4-C5, and C5-C6;
"C3-C6" encompasses C3, C4, C5, C6, C3-C6, C3-C5, C3-C4, C4-C6, C4-C5 and C5-C6 ;
"C4-C6" encompasses C4, C5, Ce, C4-C6, C4-C5 and Cs-Ce;
"C3-C6" encompasses C3, C4, C5, C6, C3-C6, C3-C5, C3-C4, C4-C6, C4-C5, and C5-C6.
As used herein, the term "leaving group" means an atom or a group of atoms that is displaced in a chemical reaction as stable species taking with it the bonding electrons. In particular, such a leaving group is selected from the group comprising: halide, in particular fluoride, chloride, bromide or iodide, (methylsulfonyl)oxy, [(trifluoromethyl)sulfonyl]oxy, [(nonafluorobutyl)sulfonyl]oxy, (phenylsulfonyl)oxy, [(4-methylphenyl)sulfonyl]oxy, [(4-bromophenyl)sulfonyl]oxy, [(4-nitrophenyl)sulfonyl]- oxy, [(2-nitrophenyl)sulfonyl]oxy, [(4-isopropylphenyl)sulfonyl]oxy, [(2,4,6-triisopropyl- phenyl)sulfonyl]oxy, [(2,4,6-trimethylphenyl)sulfonyl]oxy, [(4-ferf-butylphenyl)sulfonyl]- oxy and [(4-methoxyphenyl)sulfonyl]oxy.
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.
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".
It is possible for the compounds of general formula (I) to exist as isotopic variants. The invention therefore includes one or more isotopic variant(s) of the compounds of general formula (I), particularly deuterium-containing compounds of general formula (I). The term "Isotopic variant" of a compound or a reagent is defined as a compound exhibiting an unnatural proportion of one or more of the isotopes that constitute such a compound.
The term "Isotopic variant of the compound of general formula (I)" is defined as a compound of general formula (I) exhibiting an unnatural proportion of one or more of the isotopes that constitute such a compound.
The expression "unnatural proportion" means a proportion of such isotope which is higher than its natural abundance. The natural abundances of isotopes to be applied in this context are described in "Isotopic Compositions of the Elements 1997", Pure Appl. Chem., 70(1 ), 217-235, 1998.
Examples of such isotopes include stable and radioactive isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine and iodine, such as 2H (deuterium), 3H (tritium), 11C, 13C, 14C, 15N, 170, 180, 32P, 33P, 33S, 34S, 35S, 36S, 18F, 36CI, 82Br, 123l, 124l, 125l, 129l and 1311, respectively.
With respect to the treatment and/or prophylaxis of the disorders specified herein the isotopic variant(s) of the compounds of general formula (I) preferably contain deuterium ("deuterium-containing compounds of general formula (I)"). Isotopic variants of the compounds of general formula (I) in which one or more radioactive isotopes, such as 3H or 14C, are incorporated are useful e.g. in drug and/or substrate tissue distribution studies. These isotopes are particularly preferred for the ease of their incorporation and detectability. Positron emitting isotopes such as 18F or 11C may be incorporated into a compound of general formula (I). These isotopic variants of the compounds of general formula (I) are useful for in vivo imaging applications. Deuterium-containing and 13C- containing compounds of general formula (I) can be used in mass spectrometry analyses in the context of preclinical or clinical studies.
Isotopic variants of the compounds of general formula (I) can generally be prepared by methods known to a person skilled in the art, such as those described in the schemes and/or examples herein, by substituting a reagent for an isotopic variant of said reagent, preferably for a deuterium-containing reagent. Depending on the desired sites of deuteration, in some cases deuterium from D20 can be incorporated either directly into the compounds or into reagents that are useful for synthesizing such compounds. Deuterium gas is also a useful reagent for incorporating deuterium into molecules. Catalytic deuteration of olefinic bonds and acetylenic bonds is a rapid route for incorporation of deuterium. Metal catalysts (i.e. Pd, Pt, and Rh) in the presence of deuterium gas can be used to directly exchange deuterium for hydrogen in functional groups containing hydrocarbons. A variety of deuterated reagents and synthetic building blocks are commercially available from companies such as for example C/D/N Isotopes, Quebec, Canada; Cambridge Isotope Laboratories Inc., Andover, MA, USA; and CombiPhos Catalysts, Inc., Princeton, NJ, USA.
The term "deuterium-containing compound of general formula (I)" is defined as a compound of general formula (I), in which one or more hydrogen atom(s) is/are replaced by one or more deuterium atom(s) and in which the abundance of deuterium at each deuterated position of the compound of general formula (I) is higher than the natural abundance of deuterium, which is about 0.015%. Particularly, in a deuterium- containing compound of general formula (I) the abundance of deuterium at each deuterated position of the compound of general formula (I) is higher than 10%, 20%, 30%, 40%, 50%, 60%, 70% or 80%, preferably higher than 90%, 95%, 96% or 97%, even more preferably higher than 98% or 99% at said position(s). It is understood that the abundance of deuterium at each deuterated position is independent of the abundance of deuterium at other deuterated position(s).
The selective incorporation of one or more deuterium atom(s) into a compound of general formula (I) may alter the physicochemical properties (such as for example acidity [C. L. Perrin, et al., J. Am. Chem. Soc., 2007, 129, 4490], basicity [C. L. Perrin et al., J. Am. Chem. Soc, 2005, 127, 9641 ], lipophilicity [B. Testa et al., Int. J. Pharm., 1984, 19(3), 271 ]) and/or the metabolic profile of the molecule and may result in changes in the ratio of parent compound to metabolites or in the amounts of metabolites formed. Such changes may result in certain therapeutic advantages and hence may be preferred in some circumstances. Reduced rates of metabolism and metabolic switching, where the ratio of metabolites is changed, have been reported (A. E. Mutlib et al., Toxicol. Appl. Pharmacol., 2000, 169, 102). These changes in the exposure to parent drug and metabolites can have important consequences with respect to the pharmacodynamics, tolerability and efficacy of a deuterium-containing compound of general formula (I). In some cases deuterium substitution reduces or eliminates the formation of an undesired or toxic metabolite and enhances the formation of a desired metabolite (e.g. Nevirapine: A. M. Sharma et al., Chem. Res. Toxicol., 2013, 26, 410; Efavirenz: A. E. Mutlib et al., Toxicol. Appl. Pharmacol., 2000, 169, 102). In other cases the major effect of deuteration is to reduce the rate of systemic clearance. As a result, the biological half-life of the compound is increased. The potential clinical benefits would include the ability to maintain similar systemic exposure with decreased peak levels and increased trough levels. This could result in lower side effects and enhanced efficacy, depending on the particular compound's pharmacokinetic/ pharmacodynamic relationship. ML-337 (C. J. Wenthur et al., J. Med. Chem., 2013, 56, 5208) and Odanacatib (K. Kassahun et al., WO2012/1 12363) are examples for this deuterium effect. Still other cases have been reported in which reduced rates of metabolism result in an increase in exposure of the drug without changing the rate of systemic clearance (e.g. Rofecoxib: F. Schneider et al., Arzneim. Forsch. / Drug. Res., 2006, 56, 295; Telaprevir: F. Maltais et al., J. Med. Chem., 2009, 52, 7993). Deuterated drugs showing this effect may have reduced dosing requirements (e.g. lower number of doses or lower dosage to achieve the desired effect) and/or may produce lower metabolite loads.
A compound of general formula (I) may have multiple potential sites of attack for metabolism. To optimize the above-described effects on physicochemical properties and metabolic profile, deuterium-containing compounds of general formula (I) having a certain pattern of one or more deuterium-hydrogen exchange(s) can be selected. Particularly, the deuterium atom(s) of deuterium-containing compound(s) of general formula (I) is/are attached to a carbon atom and/or is/are located at those positions of the compound of general formula (I), which are sites of attack for metabolizing enzymes such as e.g. cytochrome P450.
In another embodiment the present invention concerns a deuterium-containing compound of general formula (I) having 1 , 2, 3 or 4 deuterium atoms, particularly with 1 , 2 or 3 deuterium atoms.
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 the present invention optionally contain one or more asymmetric centres, depending upon the location and nature of the various substituents desired. It is possible that one or more asymmetric carbon atoms are present in the (R) or (S) configuration, which can result in racemic mixtures in the case of a single asymmetric centre, and in diastereomeric mixtures in the case of multiple asymmetric centres. In certain instances, it is possible that asymmetry 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.
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, diacetyltartaric, 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 l UPAC 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.
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, see S. M. Berge, et 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, bisulfuric, 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 HCI", "x CF3COOH", "x Na+", for example, are to be understood as not a stoichiometric specification, but solely as a salt form.
The salts include water-insoluble and, particularly, water-soluble salts.
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.
Furthermore, derivatives of the compounds of formula (I) and the salts thereof which are converted into a compound of formula (I) or a salt thereof in a biological system (bioprecursors or pro-drugs) are covered by the invention. Said biological system is e.g. a mammalian organism, particularly a human subject. The bioprecursor is, for example, converted into the compound of formula (I) or a salt thereof by metabolic processes.
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, Ci-C6 alkoxymethyl esters, e.g. methoxymethyl, Ci -C6 alkanoyloxymethyl esters, e.g. pivaloyloxymethyl, phthalidyl esters, C3-C8 cycloalkoxy-carbonyloxy-Ci-Ce alkyl esters, e.g. 1 - cyclohexylcarbonyloxyethyl ; 1 ,3-dioxolen-2-onylmethyl esters, e.g. 5-methyl-1 ,3- dioxolen-2-onylmethyl ; and Ci-C6-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 the context of the properties of the compounds of the present invention the term "pharmacokinetic profile" means one single parameter or a combination thereof including permeability, bioavailability, exposure, and pharmacodynamic parameters such as duration, or magnitude of pharmacological effect, as measured in a suitable experiment. Compounds with improved pharmacokinetic profiles can, for example, be used in lower doses to achieve the same effect, may achieve a longer duration of action, or a may achieve a combination of both effects.
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. Any such combination of a compound of formula (I) of the present invention with an anticancer agent as defined below is an embodiment of the invention.
The term "(chemotherapeutic) anti-cancer agents", includes but is not limited to :
131 1-chTNT, abarelix, abiraterone, aclarubicin, adalimumab, 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, atezolizumab, 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, carbamazepine 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 alfa, dabrafenib, dasatinib, daunorubicin, decitabine, degarelix, denileukin 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, levamisole, levonorgestrel, levothyroxine sodium, lisuride, lobaplatin, lomustine, lonidamine, masoprocol, medroxyprogesterone, megestrol, melarsoprol, melphalan, mepitiostane, mercaptopurine, mesna, methadone, methotrexate, methoxsalen, methylaminolevulinate, methylprednisolone, methyltestosterone, 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, nivolumab, pentetreotide, nilotinib, nilutamide, nimorazole, nimotuzumab, nimustine, nintedanib, nitracrine, nivolumab, obinutuzumab, octreotide, ofatumumab, olaparib, olaratumab, 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, pembrolizumab, 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 , rucaparib, 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 alfa, 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.
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 Bub1 kinase 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 is mediated by Bub1 kinase, 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 intermediates used for the synthesis of the compounds of claims 1 -6 as described below, as well as their use for the synthesis of the compounds of claims 1 -6, are one further aspect of the present invention. Preferred intermediates are the Intermediate Examples as disclosed below. General Procedures
The compounds according to the invention can be prepared according to the following schemes 1 through 9.
The schemes and procedures described below illustrate synthetic routes to the compounds of general formula (I) of the invention and are not intended to be limiting. It is obvious to the person skilled in the art that the order of transformations as exemplified in the Schemes can be modified in various ways. The order of transformations exemplified in the Schemes is therefore not intended to be limiting. In addition, interconversion of any of the substituents, R\ R2, R3, R4, R7, R8, V, W, Y or Z 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, 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.
One route for the preparation of compounds of general formula (I) is described in Scheme 1 .
Scheme 1
Figure imgf000050_0001
(I)
Scheme 1 : Route for the preparation of compounds of general formula (I), wherein R1 , R2, R3, R4, R7, R8, V, W, Y and Z have the meaning as given for general formula (I), supra, X1 represents F, CI, Br, I or a sulfonate, e.g. trifluormethylsulfonate or p- toluolsulfonate. X2 represents F, CI, Br, I, boronic acid or a boronic acid ester, such as for example 4, 4, 5, 5-tetramethyl-2-phenyl-1 ,3,2-dioxaborolane (boronic acid pinacole ester). ln addition, interconversion of any of the substituents R1 , R2 R3, R4, R7, R8, V, W, Y or Z 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, 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.
Compounds of general formulae (1 -1 ) and (1 -2) are either commercially available (i.e. CAS Reg. Nos. 916325-83-2, 1363381 -84-3, 1260891 -66-4, 1060794-95-7, 121 1479- 06-9), or can be prepared according to procedures available from the public domain, as understandable to the person skilled in the art. Specific examples are described in the subsequent paragraphs.
Compounds of general formulae (1 -6) and (1 -8) are either commercially available or can be prepared according to procedures available from the public domain, as understandable to the person skilled in the art. Specific examples are described in the subsequent paragraphs.
A suitably substituted 1 H-pyrazole-3-carboxylic acid of the general formula (1 -1 ) can be reacted with methanol or ethanol in the presence of catalytic amounts of a Broensted acid, such as, for example, hydrochloric acid or sulphuric acid, at temperatures ranging from 0Ό to boiling point of the respective alcohol , preferably the reaction is carried out at 85 Ό, to furnish alkyl 1 H-indazole-3-carboxylat e intermediates of general formula (1 -2).
Alkyl 1 H-pyrazole-3-carboxylate Intermediates of the general formula (1 -2) can be converted to intermediates of general formula (1 -4) by reaction with a suitable alkylating agent, such as, for example a substituted benzyl halide (1 -3), in the presence of a suitable base, such as, for example sodium hydride, in a suitable solvent system, such as, for example, DMF, at a temperature between - 20 Ό and boiling point of the respective solvent, preferably the reaction is carried out at 0 Ό. Intermediates of general formula (1 -4) are treated with the reagent methylchloroaluminiumamide prepared in situ by addition of ammonium chloride to commercially available trimethylaluminium, in a suitable solvent system, such as, for example, toluene, at a temperature between 0Ό and the boiling point of the respective solvent, preferably the reaction is carried out at 80 Ό and are quenched with a suitable solvent system, such as, for example, methanol, to form the desired intermediate of general formula (1 -5).
Intermediates of general formula (1 -5) can be converted to intermediates of general formula (1 -7) by reaction with a suitably substituted 3,3-bis- (dimethylamino)propanenitrile of the general formula (1 -6), such as, for example 3,3- bis(dimethylamino)-2-methoxypropanenitrile, in the presence of a suitable base, such as, for example piperidine, in a suitable solvent system, such as, for example, 3- methylbutan-1 -ol, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 100Ό.
Intermediates of general formula (1 -7) can be reacted with a suitable six membered heterocycle of the general formula (1 -8), such as, for example 4-bromo-2-methyl- pyridine, in the presence of a suitable base, such as, for example sodium 2- methylpropan-2-olate, and a suitable palladium catalyst, such as for example (1 E,4£)- 1 ,5-diphenylpenta-1 ,4-dien-3-one-palladium, in the presence of a suitable ligand, such as for example 1 '-binaphthalene-2,2'-diylbis(diphenylphosphane), in a suitable solvent system, such as, for example, DMF, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at at 100Ό to furnish compounds of general formula (I). Alternatively the following palladium catalysts can be used:
allylpalladium chloride dimmer, dichlorobis(benzonitrile)palladium (II), palladium (II) acetate, palladium (II) chloride, tetrakis(triphenylphosphine)palladium (0), tris(dibenzylideneacetone)dipalladium (0), chloro(2'-amino-1 ,1 '-biphenyl-2- yl)palladium(ll) dimer, (2'-amino-1 ,1 '-biphenyl-2-yl)methanesulfonatopalladium(ll) dimer, trans-di(μ-acetato)bis[o-(di-o-tolylphosphino)benzyl]dipalladium(l l) [cataCXium® C], allylchloro[1 ,3-bis(2,4,6-trimethylphenyl)imidazol-2-ylidene]palladium(ll), allylchloro[1 ,3- bis(2,6-diisopropylphenyl)imidazol-2-ylidene]palladium(ll), chloro[(1 ,3-dimesitylimidazol- [1 ,3-bis(2,4,6-trimethylphenyl)-1 ,3-dihydro-2H-imidazol-2-ylidene](chloro){2- [(dimethylamino)methyl]phenyl}palladium, chloro[(1 ,2,3-N)-3-phenyl-2-propenyl][1 ,3- bis(2,6-di-iso-propylphenyl)imidazol-2-ylidene]palladium(ll), [2-(acetylamino)phenyl]{1 ,3- bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H-imidazol-2-ylidene}chloropalladium, {1 ,3- bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H-imidazol-2-ylidene}(chloro){2- [(dimethylamino)methyl]phenyl} palladium, {1 ,3-bis[2,6-di(propan-2-yl)phenyl]-2,3- dihydro-1 H-imidazol-2-yl}(dichloro)(3-chloropyridine-kappaN)palladium, [1 ,3-bis(2,6- diisopropylphenyl) imidazol-2-ylidene](3-chloropyridyl)palladium(ll) dichloride, [2- (acetylamino)-4-methoxyphenyl]{1 ,3-bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H- imidazol-2-ylidene}chloropalladium, {1 ,3-bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H- imidazol-2-ylidene}(chloro){2-[(dimethylamino)m
dichloro[1 ,3-bis(2,6-di-3-pentylphenyl)imidazol-2-ylidene](3-chloropyridyl) palladium(ll), dichloro(di^-chloro)bis[1 ,3-bis(2,6-di-iso-propylphenyl) imidazol-2- ylidene]dipalladium(ll), 2-(2'-di-tert-butylphosphine)biphenylpalladium(ll) acetate, chloro[dicyclohexyl(2',6'-dimethoxybiphenyl-2-yl)-lambda5-phosphanyl][2-(phenyl- kappaC2)ethanaminato-kappaN]palladium, [2-(2-aminoethyl)phenyl](chloro)palladium - di-tert-butyl[2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane, {dicyclohexyl[2',4',6'- tri(propan-2-yl)biphenyl-2-yl]phosphane}{2-[2-(methylazanidyl-kappaN)ethyl]phenyl- kappaC1 }palladium, chloro(2-dicyclohexylphosphino-2',6'-dimethoxy-1 ,1 '-biphenyl)(2'- amino-1 ,1 '-biphenyl-2-yl) palladium(ll), [2',6'-bis(propan-2-yloxy)biphenyl-2- yl](dicyclohexyl)phosphane - [2-(2-aminoethyl)phenyl](chloro)palladium, [2-(2- aminoethyl)phenyl](chloro){dicyclohexyl[2\4',6'-tri(propan-2-yl)biphenyl-2-yl]-lambda5- phosphanylidenejpalladium, 2'-(dicyclohexylphosphanyl)-N,N,N',N'-tetramethylbiphenyl- 2,6-diamine - (2'-aminobiphenyl-2-yl)(chloro)palladium, chloro(2-dicyclohexylphosphino- 2',6'-di-iso-propoxy-1 ,1 '-biphenyl)(2-amino-1 ,1 '-biphenyl-2-yl)palladium(ll), [2'-(azanidyl- kappaN)biphenyl-2-yl-kappaC2](chloro){dicyclohexyl [2',4',6'-tri(propan-2-yl)biphenyl-2- yl]-lambda5-phosphanyl}palladium, (2'-aminobi-phenyl-2-yl)(methanesulfonato- kappaO)palladium - di-tert-butyl[2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane, (2'- aminobiphenyl-2-yl)palladium(1 +) methanesulfonate - di-tert-butyl[2',4',6'-tri(propan-2- yl)biphenyl-2-yl]phosphane, dicyclohexyl[3,6-dimethoxy-2',4',6'-tri(propan-2-yl)biphenyl- 2-yl]phosphane - [2-(2-aminoethyl) phenyl](chloro)palladium, (2'-aminobiphenyl-2- yl)palladium(1 +) methanesulfonate - 2'-(dicyclohexylphosphanyl)-N,N,N',N'- tetramethylbiphenyl-2,6-diamine, sodium 2'-(dicyclohexylphosphanyl)-2,6- dimethoxybiphenyl-3-sulfonate - (2'-aminobiphenyl-2-yl)(chloro)palladium, chloro(2- dicyclohexylphosphino-2',4',6'-tri-iso-propyl-1 ,1 '-biphenyl)[2-(2- aminoethyl)phenyl]palladium(ll), (2'-aminobiphenyl-2-yl)(methane-sulfonato- kappaO)palladium - [2',6'-bis(propan-2-yloxy)biphenyl-2-yl](dicyclohexyl) phosphane, (2'-aminobiphenyl-2-yl)(methanesulfonato-kappaO)palladium - dicyclohexyl[2',4',6'- tri(propan-2-yl)biphenyl-2-yl]phosphane, (2'-aminobiphenyl-2-yl)palladium(1 +) methanesulfonate - dicyclohexyl[2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane, dicyclohexyl[3,6-dimethoxy-2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane - (2'- aminobiphenyl-2-yl)(chloro)palladium, (2'-aminobiphenyl-2-yl)(methanesulfonato- kappaO)palladium - di-tert-butyl[3,6-dimethoxy-2',4',6'-tri(propan-2-yl)biphenyl-2- yl]phosphane, (2'-aminobiphenyl-2-yl)(methanesulfonato-kappaO)palladium dicyclohexyl[3,6-dimethoxy-2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane or the following ligands:
racemic-2,2'-bis(diphenylphosphino)-1 ,1 '-binaphthyl, rac-BINAP, 1 ,1 '-bis(diphenyl- phosphino)ferrocene, bis(2-diphenylphosphinophenyl)ether, di-ferf-butylmethylphos- phonium tetrafluoroborate, 2-(di-fert-butylphosphino)biphenyl, tri-fert-butylphosphonium tetrafluoroborate, tri-2-furylphosphine, tris(2,4-di-ferf-butylphenyl)phosphite, tri-o- tolylphosphine, (9,9-dimethyl-9/-/-xanthene-4,5-diyl)bis(diphenylphosphine), dicyclohexyl(2',4',6'-triisopropyl-3,6-dimethoxybiphenyl-2-yl)phosphine, di-tert-butyl (2',4',6'-triisopropyl-3,6-dimethoxybiphenyl-2-yl)phosphine, di-tert-butyl(2',4',6'-triiso propylbiphenyl-2-yl)phosphine, dicyclohexyl(2',4',6'-triisopropylbiphenyl-2-yl) phosphine, di-tert-butyl(2',4',6'-triisopropyl-3-methoxy-6-methylbiphenyl-2-yl)phos-phine, di-tert- butyl(2',4',6'-triisopropyl-3,4,5,6-tetramethylbiphenyl-2-yl) phosphine, adamantan-1 - yl(adamantan-2-yl)(2',4',6'-triisopropyl-3,6-dimethoxybiphenyl-2-yl) phosphine, dicyclohexyl(2',6'-dimethoxybiphenyl-2-yl)phosphine, dicyclohexyl(2',6'- diisopropoxybiphenyl-2-yl)phosphine, 2'-(dicyclohexylphosphino)-N,N-dimethyl- biphenyl-2-amine, 2'-(di-tert-butylphosphino)-N,N-dimethylbiphenyl-2-amine, 2'-(di- phenylphosphino)-N,N,N',N'-tetramethylbiphenyl-2,6-diamine, di-tert-butyl(2',4',6'- tricyclohexyl-3,6-dimethoxybiphenyl-2-yl)phosphine, bis[3,5-bis(trifluoromethyl)phe-nyl] (2',4',6'-triisopropyl-3,6-dimethoxybiphenyl-2-yl)phosphine, biphenyl-2-yl(di-tert- butyl)phosphine, dicyclohexyl(2'-methylbiphenyl-2-yl)phosphine, biphenyl-2-yl (dicyclohexyl)phosphine, 2'-(dicyclohexylphosphino)-N,N-dimethylbiphenyl-2-amine, 2'- (dicyclohexylphosphino)-N,N,N',N'-tetramethylbiphenyl-2,6-diamine, sodium 2'- (dicyclohexylphosphino)-2,6-diisopropylbiphenyl-4-sulfonate, sodium 2'-
(dicyclohexylphosphino)-2,6-dimethoxybiphenyl-3-sulfonate, 1 , 1 '-binaphthalen-2-yl(di- tert-butyl)phosphine, 1 ,3-bis(2,4,6-trimethylphenyl)-1 ,3-dihydro-2H-imidazol-2-ylidene, 1 ,3-bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H-imidazol-2-ylidene.
Alternatively intermediates of general formula (1 -7) can be reacted with a suitable boronic acid or boronic acid pinacole ester of general formula (1 -8), such as, for example (2-fluoropyridin-4-yl)boronic acid, in the presence of a suitable base, such as, for example triethylamine, a suitable activating agent such as for example N,N- dimethylpyridin-4-amine and a suitable copper salt, such as for example copper (II) acetate, in a suitable solvent system, such as, for example, trichloromethane, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at room temperature to furnish compounds of general formula (I).
Alternatively intermediates of general formula (1 -7) can be reacted with a suitable six membered heterocycle of the general formula (1 -8), such as for example 4-fluoro-2- methyl-pyridine, in the presence of a suitable base, such as, for example sodium hydride, in a suitable solvent system, such as, for example DMF, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 90 Ό to furnish com pounds of general formula (I).
Compound (1 -9) can be converted into compounds of general formula (1 -6) according to the procedure depicted in Scheme 2.
Scheme 2
Figure imgf000055_0001
1 -9 1 -6
Scheme 2: Process for the transformation of compound (1 -9) into compounds of general formula (1 -6), wherein R4 has the meaning as given for general formula (I), supra.
Compound (1 -9) can be converted into compounds of general formula (1 -6) by reaction with a suitable substituted acetonitlrile derivative of the general formula (1 -10), such as, for example methoxyacetonitrile, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 80Ό.
Compounds of general formula (1 -1 1 ), where R3 is 0-(C2-C6-alkyl)OR9, can be converted into compounds of general formula (1 -13) according to the procedure depicted in Scheme 3.
Scheme 3
Figure imgf000056_0001
Scheme 3: Process for the transformation of compounds of general formula (1 -1 1 ) into compounds of general formula (1 -13), wherein R1 and R2 have the meaning as given for general formula (I), supra R12 represents a methyl- or an ethyl group or an alcohol protecting group, such as for example fert-butyldimethylsilyl, fert-butyldiphenylsilyl, triethylsilyl, triisopropylsilyl or tetrahydropyranyl and X1 represents F, CI, Br, I or a sulfonate, e.g. trifluormethylsulfonate or p-toluolsulfonate.
Compounds of general formula (1 -1 1 ) can be converted into compounds of general formula (1 -12) by reaction with a suitable reducing agent, such as, for example borane, in a suitable solvent system, such as, for example, tetrahydrofuran, in a temperature range from - 78 Ό to boiling point of the respecti ve solvent, preferably the reaction is carried out at room temperature. Compounds of general formula (1 -12) can be converted into compounds of general formula (1 -13) by reaction with a suitable halogenation or sulfonylation agent, such as for example hydrogen bromide, in a suitable solvent, such as, for example, acetic acid, in a temperature range from 0 "C to the boiling poi nt of the respective solvent, preferably the reaction is carried out at room temperature.
Compounds of general formula (1 -14) can be converted into compounds of general formula (1 -13) according to the procedure depicted in Scheme 4. Scheme 4
Figure imgf000057_0001
1-14 1-12 1-13
Scheme 4: Process for the transformation of compounds of general formula (1 -14) into compounds of general formula (1 -13), wherein R1 and R2 have the meaning as given for general formula (I), R12 represents a methyl- or an ethyl group or an alcohol protecting group, such as for example fert-butyldimethylsilyl, fert-butyldiphenylsilyl, triethylsilyl, triisopropylsilyl or tetrahydropyranyl, X1 and X3 represent F, CI, Br, I or a sulfonate, e.g. trifluormethylsulfonate or p-toluolsulfonate.
Compounds of general formula (1 -14) are reacted with a compound of general formula (1 -15) as mentioned above, in a suitable solvent, such as, for example, DMF, in the presence of a suitable base, such as, for example, sodium hydride in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at room temperature, to furnish compounds of general formula (1 -12). Compounds of general formula (1 -12) can be converted into compounds of general formula (1 -13) by reaction with a suitable halogenation or sulfonylation agent, such as for example hydrogen bromide, in a suitable solvent, such as, for example, acetic acid, in a temperature range from 0 "C to the boiling poi nt of the respective solvent, preferably the reaction is carried out at room temperature.
One route for the preparation of compounds of general formula (la), wherein R3 represents a group 0-(C2-C6-alkyl)-OH is described in Scheme 5.
Scheme 5
Figure imgf000058_0001
Figure imgf000058_0002
1-19
(la)
Scheme 5: Route for the preparation of compounds of general formula (la), wherein R1 , R2, R4, R7, R8, V, W, Y and Z have the meaning as given for general formula (I), supra, R12 represents a methyl- or an ethyl group or an alcohol protecting group, such as for example fert-butyldimethylsilyl, fert-butyldiphenylsilyl, triethylsilyl, triisopropylsilyl or tetrahydropyranyl, X1 represents F, CI, Br, I or a sulfonate, e.g. trifluormethylsulfonate or p-toluolsulfonate. X2 represents F, CI, Br, I, boronic acid or a boronic acid ester, such as for example 4, 4, 5, 5-tetramethyl-2-phenyl-1 ,3,2-dioxaborolane (boronic acid pinacole ester).
In addition, interconversion of any of the substituents R1 , R2, R4, R7, R8, V, W, Y or Z 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, 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.
Compounds of general formulae (1 -6) and (1 -8) are either commercially available or can be prepared according to procedures available from the public domain, as understandable to the person skilled in the art. Specific examples are described in the subsequent paragraphs.
Alkyl 1 H-pyrazole-3-carboxylate Intermediates of the general formula (1 -2) can be converted to intermediates of general formula (1 -16) by reaction with a suitable alkylating agent, such as, for example a substituted benzyl halide (1 -13), in the presence of a suitable base, such as, for example sodium hydride, in a suitable solvent system, such as, for example, DMF, at a temperature between - 20 Ό and boiling point of the respective solvent, preferably the reaction is carried out at 0 Ό.
Intermediates of general formula (1 -16) are treated with the reagent methylchloroaluminiumamide prepared in situ by addition of ammonium chloride to commercially available trimethylaluminium, in a suitable solvent system, such as, for example, toluene, at a temperature between 0Ό and the boiling point of the respective solvent, preferably the reaction is carried out at 80 Ό and are quenched with a suitable solvent system, such as, for example, methanol, to form the desired intermediate of general formula (1 -17).
Intermediates of general formula (1 -17) can be converted to intermediates of general formula (1 -18) by reaction with a suitably substituted 3,3-bis- (dimethylamino)propanenitrile of the general formula (1 -6), such as, for example 3,3- bis(dimethylamino)-2-methoxypropanenitrile, in the presence of a suitable base, such as, for example piperidine, in a suitable solvent system, such as, for example, 3- methylbutan-1 -ol, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 100Ό.
Intermediates of general formula (1 -18) can be reacted with a suitable six membered heterocycle of the general formula (1 -8), such as, for example 4-bromo-2-methyl- pyridine, in the presence of a suitable base, such as, for example sodium 2- methylpropan-2-olate, and a suitable palladium catalyst, such as for example (1 E,4£)- 1 ,5-diphenylpenta-1 ,4-dien-3-one-palladium, in the presence of a suitable ligand, such as for example 1 '-binaphthalene-2,2'-diylbis(diphenylphosphane), in a suitable solvent system, such as, for example, DMF, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at at 100Ό to furnish compounds of general formula (1 -19 ). Alternatively the following palladium catalysts can be used:
allylpalladium chloride dimmer, dichlorobis(benzonitrile)palladium (II), palladium (II) acetate, palladium (II) chloride, tetrakis(triphenylphosphine)palladium (0), tris(dibenzylideneacetone)dipalladium (0), chloro(2'-amino-1 ,1 '-biphenyl-2- yl)palladium(ll) dimer, (2'-amino-1 ,1 '-biphenyl-2-yl)methanesulfonatopalladium(ll) dimer, trans-di(μ-acetato)bis[o-(di-o-tolylphosphino)benzyl]dipalladium(ll) [cataCXium® C], allylchloro[1 ,3-bis(2,4,6-trimethylphenyl)imidazol-2-ylidene]palladium(ll), allylchloro[1 ,3- bis(2,6-diisopropylphenyl)imidazol-2-ylidene]palladium(ll), chloro[(1 ,3-dimesitylimidazol- [1 ,3-bis(2,4,6-trimethylphenyl)-1 ,3-dihydro-2H-imidazol-2-ylidene](chloro){2- [(dimethylamino)methyl]phenyl}palladium, chloro[(1 ,2,3-N)-3-phenyl-2-propenyl][1 ,3- bis(2,6-di-iso-propylphenyl)imidazol-2-ylidene]palladium(ll), [2-(acetylamino)phenyl]{1 ,3- bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H-imidazol-2-ylidene}chloropalladium, {1 ,3- bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H-imidazol-2-ylidene}(chloro){2- [(dimethylamino)methyl]phenyl} palladium, {1 ,3-bis[2,6-di(propan-2-yl)phenyl]-2,3- dihydro-1 H-imidazol-2-yl}(dichloro)(3-chloropyridine-kappaN)palladium, [1 ,3-bis(2,6- diisopropylphenyl) imidazol-2-ylidene](3-chloropyridyl)palladium(ll) dichloride, [2- (acetylamino)-4-methoxyphenyl]{1 ,3-bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H- imidazol-2-ylidene}chloropalladium, {1 ,3-bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H- imidazol-2-ylidene}(chloro){2-[(dimethylamino)methyl]-3,5-dimethoxyphenyl}palladium, dichloro[1 ,3-bis(2,6-di-3-pentylphenyl)imidazol-2-ylidene](3-chloropyridyl) palladium(ll), dichloro(di^-chloro)bis[1 ,3-bis(2,6-di-iso-propylphenyl) imidazol-2- ylidene]dipalladium(ll), 2-(2'-di-tert-butylphosphine)biphenylpalladium(ll) acetate, chloro[dicyclohexyl(2',6'-dimethoxybiphenyl-2-yl)-lambda5-phosphanyl][2-(phenyl- kappaC2)ethanaminato-kappaN]palladium, [2-(2-aminoethyl)phenyl](chloro)palladium - di-tert-butyl[2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane, {dicyclohexyl[2',4',6'- tri(propan-2-yl)biphenyl-2-yl]phosphane}{2-[2-(methylazanidyl-kappaN)ethyl]phenyl- kappaC1 }palladium, chloro(2-dicyclohexylphosphino-2',6'-dimethoxy-1 ,1 '-biphenyl)(2'- amino-1 ,1 '-biphenyl-2-yl) palladium(ll), [2',6'-bis(propan-2-yloxy)biphenyl-2- yl](dicyclohexyl)phosphane - [2-(2-aminoethyl)phenyl](chloro)palladium, [2-(2- aminoethyl)phenyl](chloro){dicyclohexyl[2',4',6'-tri(propan-2-yl)biphenyl-2-yl]-lambda5- phosphanylidenejpalladium, 2'-(dicyclohexylphosphanyl)-N,N,N',N'-tetramethylbiphenyl- 2,6-diamine - (2'-aminobiphenyl-2-yl)(chloro)palladium, chloro(2-dicyclohexylphosphino- 2',6'-di-iso-propoxy-1 ,1 '-biphenyl)(2-amino-1 ,1 '-biphenyl-2-yl)palladium(ll), [2'-(azanidyl- kappaN)biphenyl-2-yl-kappaC2](chloro){dicyclohexyl [2',4',6'-tri(propan-2-yl)biphenyl-2- yl]-lambda5-phosphanyl}palladium, (2'-aminobi-phenyl-2-yl)(methanesulfonato- kappaO)palladium - di-tert-butyl[2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane, (2'- aminobiphenyl-2-yl)palladium(1 +) methanesulfonate - di-tert-butyl[2',4',6'-tri(propan-2- yl)biphenyl-2-yl]phosphane, dicyclohexyl[3,6-dimethoxy-2',4',6'-tri(propan-2-yl)biphenyl- 2-yl]phosphane - [2-(2-aminoethyl) phenyl](chloro)palladium, (2'-aminobiphenyl-2- yl)palladium(1 +) methanesulfonate - 2'-(dicyclohexylphosphanyl)-N,N,N',N'- tetramethylbiphenyl-2,6-diamine, sodium 2'-(dicyclohexylphosphanyl)-2,6- dimethoxybiphenyl-3-sulfonate - (2'-aminobiphenyl-2-yl)(chloro)palladium, chloro(2- dicyclohexylphosphino-2',4',6'-tri-iso-propyl-1 ,1 '-biphenyl)[2-(2- aminoethyl)phenyl]palladium(ll), (2'-aminobiphenyl-2-yl)(methane-sulfonato- kappaO)palladium - [2',6'-bis(propan-2-yloxy)biphenyl-2-yl](dicyclohexyl) phosphane, (2'-aminobiphenyl-2-yl)(methanesulfonato-kappaO)palladium - dicyclohexyl[2',4',6'- tri(propan-2-yl)biphenyl-2-yl]phosphane, (2'-aminobiphenyl-2-yl)palladium(1 +) methanesulfonate - dicyclohexyl[2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane, dicyclohexyl[3,6-dimethoxy-2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane - (2'- aminobiphenyl-2-yl)(chloro)palladium, (2'-aminobiphenyl-2-yl)(methanesulfonato- kappaO)palladium - di-tert-butyl[3,6-dimethoxy-2',4',6'-tri(propan-2-yl)biphenyl-2- yljphosphane, (2'-aminobiphenyl-2-yl)(methanesulfonato-kappaO)palladium dicyclohexyl[3,6-dimethoxy-2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane or the following ligands:
racemic-2,2'-bis(diphenylphosphino)-1 ,1 '-binaphthyl, rac-BINAP, 1 ,1 '-bis(diphenyl- phosphino)ferrocene, bis(2-diphenylphosphinophenyl)ether, di-ferf-butylmethylphos- phonium tetrafluoroborate, 2-(di-fert-butylphosphino)biphenyl, tri-fert-butylphosphonium tetrafluoroborate, tri-2-furylphosphine, tris(2,4-di-ferf-butylphenyl)phosphite, tri-o- tolylphosphine, (9,9-dimethyl-9/-/-xanthene-4,5-diyl)bis(diphenylphosphine), dicyclohexyl(2',4',6'-triisopropyl-3,6-dimethoxybiphenyl-2-yl)phosphine, di-tert-butyl (2',4',6'-triisopropyl-3,6-dimethoxybiphenyl-2-yl)phosphine, di-tert-butyl(2',4',6'-triiso propylbiphenyl-2-yl)phosphine, dicyclohexyl(2',4',6'-triisopropylbiphenyl-2-yl) phosphine, di-tert-butyl(2',4',6'-triisopropyl-3-methoxy-6-methylbiphenyl-2-yl)phos-phine, di-tert- butyl(2',4',6'-triisopropyl-3,4,5,6-tetramethylbiphenyl-2-yl) phosphine, adamantan-1 - yl(adamantan-2-yl)(2',4',6'-triisopropyl-3,6-dimethoxybiphenyl-2-yl) phosphine, dicyclohexyl(2',6'-dimethoxybiphenyl-2-yl)phosphine, dicyclohexyl(2',6'- diisopropoxybiphenyl-2-yl)phosphine, 2'-(dicyclohexylphosphino)-N,N-dimethyl- biphenyl-2-amine, 2'-(di-tert-butylphosphino)-N,N-dimethylbiphenyl-2-amine, 2'-(di- phenylphosphino)-N,N,N',N'-tetramethylbiphenyl-2,6-diamine, di-tert-butyl(2',4',6'- tricyclohexyl-3,6-dimethoxybiphenyl-2-yl)phosphine, bis[3,5-bis(trifluoromethyl)phe-nyl] (2',4',6'-triisopropyl-3,6-dimethoxybiphenyl-2-yl)phosphine, biphenyl-2-yl(di-tert- butyl)phosphine, dicyclohexyl(2'-methylbiphenyl-2-yl)phosphine, biphenyl-2-yl (dicyclohexyl)phosphine, 2'-(dicyclohexylphosphino)-N,N-dimethylbiphenyl-2-amine, 2'- (dicyclohexylphosphino)-N,N,N\N'-tetramethylbiphenyl-2,6-diamine, sodium 2'- (dicyclohexylphosphino)-2,6-diisopropylbiphenyl-4-sulfonate, sodium 2'-
(dicyclohexylphosphino)-2,6-dimethoxybiphenyl-3-sulfonate, 1 , 1 '-binaphthalen-2-yl(di- tert-butyl)phosphine, 1 ,3-bis(2,4,6-trimethylphenyl)-1 ,3-dihydro-2H-imidazol-2-ylidene, 1 ,3-bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H-imidazol-2-ylidene.
Alternatively intermediates of general formula (1 -18) can be reacted with a suitable boronic acid or boronic acid pinacole ester of general formula (1 -8), such as, for example (2-fluoropyridin-4-yl)boronic acid, in the presence of a suitable base, such as, for example triethylamine, a suitable activating agent such as for example N,N- dimethylpyridin-4-amine and a suitable copper salt, such as for example copper (II) acetate, in a suitable solvent system, such as, for example, trichloromethane, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at room temperature to furnish compounds of general formula (1 -19).
Alternatively intermediates of general formula (1 -18) can be reacted with a suitable six membered heterocycle of the general formula (1 -8), such as for example 4-fluoro-2- methyl-pyridine, in the presence of a suitable base, such as, for example sodium hydride, in a suitable solvent system, such as, for example DMF, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 90 Ό to furnish com pounds of general formula (1 -19).
Compounds of general formula (1 -19) are converted to compounds of general formula (la) by treatment with a suitable deprotection agent, such as a dealkylating agent, such as for example boron trichloride, in a suitable solvent, such as, for example, dichloromethane, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at room temperature. Compounds of general formula (1 -5) can be converted into compounds of general formula (1 -7) according to the procedure depicted in Scheme 6. Scheme 6
Figure imgf000063_0001
Scheme 6: Alternative route for the preparation of compounds of general formula (1 -7), wherein R1 , R2, R3 and R4 have the meaning as given for general formula (I), supra.
In addition, interconversion of any of the substituents R1 , R2, R3 or R4 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, 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 para-graphs.
Intermediates of general formula (1 -5) can be converted to intermediates of general formula (1 -7) by reaction with a suitably substituted 3-methoxyacrylonitrile of the general formula (1 -20), such as, for example (ethoxymethylene)malononitrile, in the presence of a suitable base, such as, for example sodium methanolate, in a suitable solvent system, such as, for example, methanol, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 65Ό.
Scheme 7
Figure imgf000064_0001
1 -22 1-5
Scheme 7: Route for the preparation of compounds of general formula (1 -5), R1 , R2, R3 and R4 have the meaning as given for general formula (I), supra. R9 represents a methyl- or an ethyl group or an alcohol protecting group, such as for example tert- butyldimethylsilyl, tert-butyldiphenylsilyl, triethylsilyl, triisopropylsilyl or tetrahydropyranyl. Intermediates of general formula (1 -4) can be converted to intermediates of general formula (1 -21 ) by reaction with ammonia, in a suitable solvent system, such as, for example, methanol, at a temperature between 0 Ό an d boiling point of the respective solvent, preferably the reaction is carried out at 50 Ό, at a pressure between 1 and 10 bar, preferably the reaction is carried in a sealed vessel.
Intermediates of general formula (1 -21 ) are treated with triflic anhydride, in a suitable solvent system, such as, for example, tetrahydrofuran, in the presence of a suitable base, such as, for example, pyridine, at a temperature between 0Ό and the boiling point of the respective solvent, preferably the reaction is carried out at room temperature, to form the desired intermediate of general formula (1 -22).
Intermediates of general formula (1 -22) can be converted to intermediates of general formula (1 -5) by reaction with a suitable alcoholate, such as, for example sodium methanolate, in a suitable solvent system, such as, for example, the corresponding alcohol, e.g. methanol, at a temperature between room temperature and the boiling point of the respective solvent, preferably the reaction is carried out at room temperature, and subsequent treatment with a suitable source of ammonium, such as for example, ammonium chloride in the presence of a suitable acid, such as for example acetic acid in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 50Ό.
An alternative route for the preparation of compounds of general formula (1 -22) is described in Scheme 8.
Scheme 8
Figure imgf000065_0001
Scheme 8: Route for the preparation of compounds of general formula (1 -22), R1 , R2, R3, R7 and R8 have the meaning as given for general formula (I), supra, X1 represents F, CI, Br, I or a sulfonate, e.g. trifluormethylsulfonate or p-toluolsulfonate.
Compounds of general formula (1 -29) are either commercially available (i.e. CAS Reg. Nos. 68618-36-0, 76006-13-8, 633328-33-3, 1785345-95-0, 1351386-47-4, 1781061 - 48-0, 1780665-28-2), or can be prepared according to procedures available from the public domain, as understandable to the person skilled in the art. Specific examples are described in the subsequent paragraphs.
Intermediates of general formula (1 -29) can be converted to intermediates of general formula (1 -30) by reaction with a suitable alkylating agent, such as, for example a substituted benzyl halide (1 -3), in the presence of a suitable base, such as, for example cesium carbonate or sodium hydride, in a suitable solvent system, such as, for example, DMF, at a temperature between - 20 Ό and boiling point of the respective solvent, preferably the reaction is carried out at 0 Ό.
Intermediates of general formula (1 -30) can be converted to intermediates of general formula (1 -22) by reaction with zinc cynanide, in the presence of a catalyst, such as for example [1 ,1 '-bis(diphenylphosphino)ferrocene]dichloropalladium(ll) complex with dichloromethane, in a suitable solvent system, such as, for example, DMF, at a temperature between 50Ό and the boiling point of t he respective solvent, preferably the reaction is carried out at 120Ό.
An alternative route for the preparation of compounds of general formula (I) is described in Scheme 9.
Scheme 9
Figure imgf000067_0001
Scheme 9: Route for the preparation of compounds of general formula (I), wherein R1 , R2, R3, R4, V, W, Y and Z have the meaning as given for general formula (I), supra, X1 represents F, CI, Br, I or a sulfonate, e.g. trifluormethylsulfonate or p-toluolsulfonate, and X2 represents F, CI, Br, I, boronic acid or a boronic acid ester, such as for example 4,4,5,5-tetramethyl-2-phenyl-1 ,3,2-dioxaborolane (boronic acid pinacole ester). ln addition, interconversion of any of the substituents R1 , R2, R3, R4, V, W, Y or Z 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, 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.
Compounds 1 -3, 1 -6, 1 -8 and 1 -30 are either commercially available or can be prepared according to procedures available from the public domain, as understandable to the person skilled in the art. Specific examples are described in the subsequent paragraphs.
AlkyI 1 H-pyrazole-3-carboxylate Intermediates of the general formula (1 -2) can be converted to intermediates of general formula (1 -24) by reaction with a suitable alkylating agent, such as, for example a substituted benzyl halide (1 -23), in the presence of a suitable base, such as, for example sodium hydride, in a suitable solvent system, such as, for example, DMF, at a temperature between - 20 Ό and boiling point of the respective solvent, preferably the reaction is carried out at 0 Ό.
Intermediates of general formula (1 -24) are treated with the reagent methylchloroaluminiumamide prepared in situ by addition of ammonium chloride to commercially available trimethylaluminium, in a suitable solvent system, such as, for example, toluene, at a temperature between 0Ό and the boiling point of the respective solvent, preferably the reaction is carried out at 80 Ό and are quenched with a suitable solvent system, such as, for example, methanol, to form the desired intermediate of general formula(1 -25).
Intermediates of general formula (1 -25) can be converted to intermediates of general formula (1 -26) by reaction with a suitably substituted 3,3-bis- (dimethylamino)propanenitrile of the general formula (1 -6), such as, for example 3,3- bis(dimethylamino)-2-methoxypropanenitrile, in the presence of a suitable base, such as, for example piperidine, in a suitable solvent system, such as, for example, 3- methylbutan-1 -ol, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 100Ό.
Intermediates of general formula (1 -26) can be converted to intermediates of general formula (1 -27) by reaction with a suitably Broensted acid, such as, for example methanesulfonic acid and trifluoroacetic acid,, in a suitable solvent system, such as, for example, dichloromethane, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at room temperature.
Intermediates of the general formula (1 -27) can be converted to intermediates of general formula (1 -7) by reaction with a suitable alkylating agent, such as, for example a substituted benzyl halide (1 -3), in the presence of a suitable base, such as, for example sodium hydride, in a suitable solvent system, such as, for example, DMF, at a temperature between - 20 Ό and boiling point of th e respective solvent, preferably the reaction is carried out at 0 Ό.
Intermediates of general formula (1 -7) can be reacted with a suitable six membered heterocycle of the general formula (1 -8), such as, for example 4-bromo-2-methyl- pyridine, in the presence of a suitable base, such as, for example sodium 2- methylpropan-2-olate, and a suitable palladium catalyst, such as for example (1 E,4£)- 1 ,5-diphenylpenta-1 ,4-dien-3-one-palladium, in the presence of a suitable ligand, such as for example 1 '-binaphthalene-2,2'-diylbis(diphenylphosphane), in a suitable solvent system, such as, for example, DMF, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at at 100Ό to furnish compounds of general formula (I). Alternatively the following palladium catalysts can be used:
allylpalladium chloride dimmer, dichlorobis(benzonitrile)palladium (II), palladium (II) acetate, palladium (II) chloride, tetrakis(triphenylphosphine)palladium (0), tris(dibenzylideneacetone)dipalladium (0), chloro(2'-amino-1 ,1 '-biphenyl-2- yl)palladium(ll) dimer, (2'-amino-1 ,1 '-biphenyl-2-yl)methanesulfonatopalladium(ll) dimer, trans-di(μ-acetato)bis[o-(di-o-tolylphosphino)benzyl]dipalladium(ll) [cataCXium® C], allylchloro[1 ,3-bis(2,4,6-trimethylphenyl)imidazol-2-ylidene]palladium(ll), allylchloro[1 ,3- bis(2,6-diisopropylphenyl)imidazol-2-ylidene]palladium(ll), chloro[(1 ,3-dimesitylimidazol- [1 ,3-bis(2,4,6-trimethylphenyl)-1 ,3-dihydro-2H-imidazol-2-ylidene](chloro){2- [(dimethylamino)methyl]phenyl}palladium, chloro[(1 ,2,3-N)-3-phenyl-2-propenyl][1 ,3- bis(2,6-di-iso-propylphenyl)imidazol-2-ylidene]palladium(ll), [2-(acetylamino)phenyl]{1 ,3- bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H-imidazol-2-ylidene}chloropalladium, {1 ,3- bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H-imidazol-2-ylidene}(chloro){2- [(dimethylamino)methyl]phenyl} palladium, {1 ,3-bis[2,6-di(propan-2-yl)phenyl]-2,3- dihydro-1 H-imidazol-2-yl}(dichloro)(3-chloropyridine-kappaN)palladium, [1 ,3-bis(2,6- diisopropylphenyl) imidazol-2-ylidene](3-chloropyridyl)palladium(ll) dichloride, [2- (acetylamino)-4-methoxyphenyl]{1 ,3-bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H- imidazol-2-ylidene}chloropalladium, {1 ,3-bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H- imidazol-2-ylidene}(chloro){2-[(dimethylamino)methyl]-3,5-dimethoxyphenyl}palladium, dichloro[1 ,3-bis(2,6-di-3-pentylphenyl)imidazol-2-ylidene](3-chloropyridyl) palladium(ll), dichloro(di-μ-chloro)bis[1 ,3-bis(2,6-di-iso-propylphenyl) imidazol-2- ylidene]dipalladium(ll), 2-(2'-di-tert-butylphosphine)biphenylpalladium(ll) acetate, chloro[dicyclohexyl(2',6'-dimethoxybiphenyl-2-yl)-lambda5-phosphanyl][2-(phenyl- kappaC2)ethanaminato-kappaN]palladium, [2-(2-aminoethyl)phenyl](chloro)palladium - di-tert-butyl[2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane, {dicyclohexyl[2',4',6'- tri(propan-2-yl)biphenyl-2-yl]phosphane}{2-[2-(methylazanidyl-kappaN)ethyl]phenyl- kappaC1 }palladium, chloro(2-dicyclohexylphosphino-2',6'-dimethoxy-1 ,1 '-biphenyl)(2'- amino-1 ,1 '-biphenyl-2-yl) palladium(ll), [2',6'-bis(propan-2-yloxy)biphenyl-2- yl](dicyclohexyl)phosphane - [2-(2-aminoethyl)phenyl](chloro)palladium, [2-(2- aminoethyl)phenyl](chloro){dicyclohexyl[2',4',6'-tri(propan-2-yl)biphenyl-2-yl]-lambda5- phosphanylidenejpalladium, 2'-(dicyclohexylphosphanyl)-N,N,N',N'-tetramethylbiphenyl- 2,6-diamine - (2'-aminobiphenyl-2-yl)(chloro)palladium, chloro(2-dicyclohexylphosphino- 2',6'-di-iso-propoxy-1 ,1 '-biphenyl)(2-amino-1 ,1 '-biphenyl-2-yl)palladium(ll), [2'-(azanidyl- kappaN)biphenyl-2-yl-kappaC2](chloro){dicyclohexyl [2',4',6'-tri(propan-2-yl)biphenyl-2- yl]-lambda5-phosphanyl}palladium, (2'-aminobi-phenyl-2-yl)(methanesulfonato- kappaO)palladium - di-tert-butyl[2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane, (2'- aminobiphenyl-2-yl)palladium(1 +) methanesulfonate - di-tert-butyl[2',4',6'-tri(propan-2- yl)biphenyl-2-yl]phosphane, dicyclohexyl[3,6-dimethoxy-2',4',6'-tri(propan-2-yl)biphenyl- 2-yl]phosphane - [2-(2-aminoethyl) phenyl](chloro)palladium, (2'-aminobiphenyl-2- yl)palladium(1 +) methanesulfonate - 2'-(dicyclohexylphosphanyl)-N,N,N',N'- tetramethylbiphenyl-2,6-diamine, sodium 2'-(dicyclohexylphosphanyl)-2,6- dimethoxybiphenyl-3-sulfonate - (2'-aminobiphenyl-2-yl)(chloro)palladium, chloro(2- dicyclohexylphosphino-2',4',6'-tri-iso-propyl-1 ,1 '-biphenyl)[2-(2- aminoethyl)phenyl]palladium(ll), (2'-aminobiphenyl-2-yl)(methane-sulfonato- kappaO)palladium - [2',6'-bis(propan-2-yloxy)biphenyl-2-yl](dicyclohexyl) phosphane, (2'-aminobiphenyl-2-yl)(methanesulfonato-kappaO)palladium - dicyclohexyl[2',4',6'- tri(propan-2-yl)biphenyl-2-yl]phosphane, (2'-aminobiphenyl-2-yl)palladium(1 +) methanesulfonate - dicyclohexyl[2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane, dicyclohexyl[3,6-dimethoxy-2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane - (2'- aminobiphenyl-2-yl)(chloro)palladium, (2'-aminobiphenyl-2-yl)(methanesulfonato- kappaO)palladium - di-tert-butyl[3,6-dimethoxy-2',4',6'-tri(propan-2-yl)biphenyl-2- yl]phosphane, (2'-aminobiphenyl-2-yl)(methanesulfonato-kappaO)palladium dicyclohexyl[3,6-dimethoxy-2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane or the following ligands:
racemic-2,2'-bis(diphenylphosphino)-1 ,1 '-binaphthyl, rac-BINAP, 1 ,1 '-bis(diphenyl- phosphino)ferrocene, bis(2-diphenylphosphinophenyl)ether, di-ferf-butylmethylphos- phonium tetrafluoroborate, 2-(di-fert-butylphosphino)biphenyl, tri-fert-butylphosphonium tetrafluoroborate, tri-2-furylphosphine, tris(2,4-di-ferf-butylphenyl)phosphite, tri-o- tolylphosphine, (9,9-dimethyl-9/-/-xanthene-4,5-diyl)bis(diphenylphosphine), dicyclohexyl(2',4',6'-triisopropyl-3,6-dimethoxybiphenyl-2-yl)phosphine, di-tert-butyl (2',4',6'-triisopropyl-3,6-dimethoxybiphenyl-2-yl)phosphine, di-tert-butyl(2',4',6'-triiso propylbiphenyl-2-yl)phosphine, dicyclohexyl(2',4',6'-triisopropylbiphenyl-2-yl) phosphine, di-tert-butyl(2',4',6'-triisopropyl-3-methoxy-6-methylbiphenyl-2-yl)phos-phine, di-tert- butyl(2',4',6'-triisopropyl-3,4,5,6-tetramethylbiphenyl-2-yl) phosphine, adamantan-1 - yl(adamantan-2-yl)(2',4',6'-triisopropyl-3,6-dimethoxybiphenyl-2-yl) phosphine, dicyclohexyl(2',6'-dimethoxybiphenyl-2-yl)phosphine, dicyclohexyl(2',6'- diisopropoxybiphenyl-2-yl)phosphine, 2'-(dicyclohexylphosphino)-N,N-dimethyl- biphenyl-2-amine, 2'-(di-tert-butylphosphino)-N,N-dimethylbiphenyl-2-amine, 2'-(di- phenylphosphino)-N,N,N',N'-tetramethylbiphenyl-2,6-diamine, di-tert-butyl(2',4',6'- tricyclohexyl-3,6-dimethoxybiphenyl-2-yl)phosphine, bis[3,5-bis(trifluoromethyl)phe-nyl] (2',4',6'-triisopropyl-3,6-dimethoxybiphenyl-2-yl)phosphine, biphenyl-2-yl(di-tert- butyl)phosphine, dicyclohexyl(2'-methylbiphenyl-2-yl)phosphine, biphenyl-2-yl (dicyclohexyl)phosphine, 2'-(dicyclohexylphosphino)-N,N-dimethylbiphenyl-2-amine, 2'- (dicyclohexylphosphino)-N,N,N',N'-tetramethylbiphenyl-2,6-diamine, sodium 2'- (dicyclohexylphosphino)-2,6-diisopropylbiphenyl-4-sulfonate, sodium 2'-
(dicyclohexylphosphino)-2,6-dimethoxybiphenyl-3-sulfonate, 1 , 1 '-binaphthalen-2-yl(di- tert-butyl)phosphine, 1 ,3-bis(2,4,6-trimethylphenyl)-1 ,3-dihydro-2H-imidazol-2-ylidene, 1 ,3-bis[2,6-di(propan-2-yl)phenyl]-1 ,3-dihydro-2H-imidazol-2-ylidene.
Alternatively intermediates of general formula (1 -7) can be reacted with a suitable boronic acid or boronic acid pinacole ester of general formula (1 -8), such as, for example (2-fluoropyridin-4-yl)boronic acid, in the presence of a suitable base, such as, for example triethylamine, a suitable activating agent such as for example N,N- dimethylpyridin-4-amine and a suitable copper salt, such as for example copper (II) acetate, in a suitable solvent system, such as, for example, trichloromethane, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at room temperature to furnish compounds of general formula (I).
Alternatively intermediates of general formula (1 -7) can be reacted with a suitable six membered heterocycle of the general formula (1 -8), such as for example 4-fluoro-2- methyl-pyridine, in the presence of a suitable base, such as, for example sodium hydride, in a suitable solvent system, such as, for example DMF, in a temperature range from room temperature to the boiling point of the respective solvent, preferably the reaction is carried out at 90 Ό to furnish com pounds of general formula (I).
It is known to the person skilled in the art that, if there are a number of reactive centers on a starting or intermediate compound, it may be necessary to block one or more reactive centers temporarily by protective groups in order to allow a reaction to proceed specifically at the desired reaction center. A detailed description for the use of a large number of proven protective groups is found, for example, in T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, 1999, 3rd Ed., or in P. Kocienski, Protecting Groups, Thieme Medical Publishers, 2000.
The compounds according to the invention are isolated and purified in a manner known per se, e.g. by distilling off the solvent in vacuo and recrystallizing the residue obtained from a suitable solvent or subjecting it to one of the customary purification methods, such as chromatography on a suitable support material. Furthermore, reverse phase preparative HPLC of compounds of the present invention which possess a sufficiently basic or acidic functionality, may result in the formation of a salt, such as, in the case of a compound of the present invention which is sufficiently basic, a trifluoroacetate or formate salt for example, or, in the case of a compound of the present invention which is sufficiently acidic, an ammonium salt for example. Salts of this type can either be transformed into its free base or free acid form, respectively, by various methods known to the persion skilled in the art, or be used as salts in subsequent biological assays. Additionally, the drying process during the isolation of compounds of the present invention may not fully remove traces of cosolvents, especially such as formic acid or trifluoroacetic acid, to give solvates or inclusion complexes. The person skilled in the art will recognise which solvates or inclusion complexes are acceptable to be used in subsequent biological assays. It is to be understood that the specific form (e.g. salt, free base, solvate, inclusion complex) of a compound of the present invention as isolated as described herein is not necessarily the only form in which said compound can be applied to a biological assay in order to quantify the specific biological activity.
Salts of the compounds of formula (I) according to the invention can be obtained by dissolving the free compound in a suitable solvent (for example a ketone such as acetone, methylethylketone or methylisobutylketone, an ether such as diethyl ether, tetrahydrofuran or dioxane, a chlorinated hydrocarbon such as methylene chloride or chloroform, or a low molecular weight aliphatic alcohol such as methanol, ethanol or isopropanol) which contains the desired acid or base, or to which the desired acid or base is then added. The acid or base can be employed in salt preparation, depending on whether a mono- or polybasic acid or base is concerned and depending on which salt is desired, in an equimolar quantitative ratio or one differing therefrom. The salts are obtained by filtering, reprecipitating, precipitating with a non-solvent for the salt or by evaporating the solvent. Salts obtained can be converted into the free compounds which, in turn, can be converted into salts. In this manner, pharmaceutically unacceptable salts, which can be obtained, for example, as process products in the manufacturing on an industrial scale, can be converted into pharmaceutically acceptable salts by processes known to the person skilled in the art. Especially preferred are hydrochlorides and the process used in the examples section.
Pure diastereomers and pure enantiomers of the compounds and salts according to the invention can be obtained e.g. by asymmetric synthesis, by using chiral starting compounds in synthesis and by splitting up enantiomeric and diasteriomeric mixtures obtained in synthesis.
Enantiomeric and diastereomeric mixtures can be split up into the pure enantiomers and pure diastereomers by methods known to a person skilled in the art. Preferably, diastereomeric mixtures are separated by crystallization, in particular fractional crystallization, or chromatography. Enantiomeric mixtures can be separated e.g. by forming diastereomers with a chiral auxiliary agent, resolving the diastereomers obtained and removing the chiral auxiliary agent. As chiral auxiliary agents, for example, chiral acids can be used to separate enantiomeric bases such as e.g. mandelic acid and chiral bases can be used to separate enantiomeric acids via formation of diastereomeric salts. Furthermore, diastereomeric derivatives such as diastereomeric esters can be formed from enantiomeric mixtures of alcohols or enantiomeric mixtures of acids, respectively, using chiral acids or chiral alcohols, respectively, as chiral auxiliary agents. Additionally, diastereomeric complexes or diastereomeric clathrates may be used for separating enantiomeric mixtures. Alternatively, enantiomeric mixtures can be split up using chiral separating columns in chromatography. Another suitable method for the isolation of enantiomers is the enzymatic separation.
One preferred aspect of the invention is the process for the preparation of the compounds of claims 1 -6 according to the examples.
Optionally, compounds of the formula (I) can be converted into their salts, or, optionally, salts of the compounds of the formula (I) can be converted into the free compounds. Corresponding processes are customary for the skilled person.
Optionally, compounds of the formula (I) can be converted into their N-oxides. The N- oxide may also be introduced by way of an intermediate. N-oxides may be prepared by treating an appropriate precursor with an oxidizing agent, such as meta- chloroperbenzoic acid, in an appropriate solvent, such as DCM, at suitable temperatures, such as from 0 Ό to 40 "C, whereby r oom temperature is generally preferred. Further corresponding processes for forming N-oxides are customary for the skilled person.
One preferred aspect of the invention is the process for the preparation of the compounds of claims 1 -6 according to the examples, as well as the intermediates used for their preparation.
Optionally, compounds of the formula (I) can be converted into their salts, or, optionally, salts of the compounds of the formula (I) can be converted into the free compounds. Corresponding processes are customary for the skilled person.
Commercial utility
As mentioned supra, the compounds of the present invention have surprisingly been found to effectively inhibit Bub1 finally resulting in cell death e.g. apoptosis 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 is mediated by Bub1 , such as, for example, benign and malignant neoplasia, more specifically 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,
especially haematological tumours, solid tumours, and/or metastases of breast, bladder, bone, brain, central and peripheral nervous system, cervix, colon, endocrine glands (e.g. thyroid and adrenal cortex), endocrine tumours, endometrium, esophagus, gastrointestinal tumours, germ cells, kidney, liver, lung, larynx and hypopharynx, mesothelioma, ovary, pancreas, prostate, rectum, renal, small intestine, soft tissue, stomach, skin, testis, ureter, vagina and vulva as well as malignant neoplasias including primary tumors in said organs and corresponding secondary tumors in distant organs ("tumor metastases"). Haematological tumors can e.g be exemplified by aggressive and indolent forms of leukemia and lymphoma, namely non-Hodgkins disease, chronic and acute myeloid leukemia (CML / AML), acute lymphoblastic leukemia (ALL), Hodgkins disease, multiple myeloma and T-cell lymphoma. Also included are myelodysplastic syndrome, plasma cell neoplasia, paraneoplastic syndromes, and cancers of unknown primary site as well as AIDS related malignancies.
A further aspect of the invention is the use of the compounds according to formula (I) for the treatment of cer-vical -, breast -, non-small cell lung -, prostate -, colon - and melanoma tumors and/or metastases thereof, especially preferred for the treatment thereof as well as a method of treatment of cervical -, breast -, non-small cell lung -, prostate -, colon - and melanoma tumors and/or metastases thereof comprising administering an effective amount of a compound of formula (I). One aspect of the invention is the use of the compounds according to formula (I) for the treatment of cervix tumors as well as a method of treatment of cervix tumors comprising administering an effective amount of a compound of formula (I).
In accordance with an aspect of the present invention therefore the invention relates to a compound of general formula 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 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, especially for use in the 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 the prophylaxis or treatment of hyperproliferative disorders or disorders responsive to induction of cell death i.e apoptosis. .
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 preferably 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, especially the treatment, wherein the diseases are haematological tumours, solid tumours and/or metastases thereof.
Another aspect is the use of a compound of formula (I) is for the treatment of cervical -, breast -, non-small cell lung -, prostate -, colon - and melanoma tumors and/or metastases thereof, especially preferred for the treatment thereof. A preferred aspect is the use of a compound of formula (I) for the prophylaxis and/or treatment of cervical tumors especially preferred for the treatment thereof.
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, wherein such disease is a hyperproliferative disorder or a disorder responsive to induction of cell death e.g.apoptosis. In an embodiment the disease is a haematological tumour, a solid tumour and/or metastases thereof. In another embodiment the disease is cervical -, breast -, non-small cell lung -, prostate -, colon - and melanoma tumor and/or metastases thereof, in a preferred aspect the disease is cervical tumor.
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 cell death e.g. 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. Hyper-proliferative 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 AIDS-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 kinase disorders The present invention also provides methods for the treatment of disorders associated with aberrant mitogen extracellular kinase activity, including, but not limited to stroke, heart failure, hepatomegaly, cardiomegaly, diabetes, Alzheimer's disease, cystic fibrosis, symptoms of xenograft rejections, septic shock or asthma.
Effective amounts of compounds of the present invention can be used to treat such disorders, including those diseases (e.g., cancer) mentioned in the Background section above. Nonetheless, such cancers and other diseases can be treated with compounds of the present invention, regardless of the mechanism of action and/or the relationship between the kinase and the disorder.
The phrase "aberrant kinase activity" or "aberrant tyrosine kinase activity," includes any abnormal expression or activity of the gene encoding the kinase or of the polypeptide it encodes. Examples of such aberrant activity, include, but are not limited to, over- expression of the gene or polypeptide ; gene amplification ; mutations which produce constitutively-active or hyperactive kinase activity ; gene mutations, deletions, substitutions, additions, etc.
The present invention also provides for methods of inhibiting a kinase activity, especially of mitogen extracellular kinase, comprising administering an effective amount of a compound of the present invention, including salts, polymorphs, metabolites, hydrates, solvates, prodrugs (e.g.: esters) thereof, and diastereoisomeric forms thereof. Kinase activity can be inhibited in cells (e.g., in vitro), or in the cells of a mammalian subject, especially a human patient in need of treatment.
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 , 1480 ; 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 e.g. apoptosis of such cell types.
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, especially in therapy of tumour growth and metastases, especially in solid tumours of all indications and stages with or without pre-treatment of the tumour growth.
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 or auxiliary and a pharmaceutically effective amount of a compound, or salt thereof, of the present invention.
Another aspect of the invention is a pharmaceutical composition comprising a pharmaceutically effective amount of a compound of formula (I) and a pharmaceutically acceptable auxiliary for the treatment of a disease mentioned supra, especially for the treatment of haematological tumours, solid tumours and/or metastases thereof.
A pharmaceutically acceptable carrier or auxiliary is preferably a carrier that is 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. Carriers and auxiliaries are all kinds of additives assisting to the composition to be suitable for administration.
A pharmaceutically effective amount of compound is preferably that amount which produces a result or exerts the intended influence on the particular condition being treated.
The compounds of the present invention can be administered with pharmaceutically- acceptable carriers or auxiliaries 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 auxiliaries, 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. The suspensions may also contain one or more preservatives, for example, ethyl or n-propyl p-hydroxybenzoate ; 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 poly(ethylene 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, methycellulose, 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 quarternary 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.
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 administration, 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 & 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) ; alkalinizing 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 charcoa)l ; aerosol propellants (examples include but are not limited to carbon dioxide, CCI2F2,
Figure imgf000085_0001
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) ; leviqatinq 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 stearate 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 opaguants (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 wettinq 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 i.v. 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 i.v. infusion over about 60 minutes.
Lvophilised powder for i.v. 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 minutes.
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.
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.
Combination Therapies
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. Those combined pharmaceutical agents can be other agents having antiproliferative effects such as for example for the treatment of haematological tumours, solid tumours and/or metastases thereof and/or agents for the treatment of undesired side effects. The present invention relates also to such combinations.
Other anti-hyper-proliferative agents suitable for use with the composition of the invention include but are not limited to those compounds acknowledged to be used in the treatment of neoplastic diseases in Goodman and Gilman's The Pharmacological Basis of Therapeutics (Ninth Edition), editor Molinoff et al., publ. by McGraw-Hill, pages 1225-1287, (1996), which is hereby incorporated by reference, especially (chemotherapeutic) anti-cancer agents as defined supra. The combination can be a non-fixed combination or a fixed-dose combination as the case may be. 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.
As will be appreciated by persons skilled in the art, the invention is not limited to the particular embodiments described herein, but covers all modifications of said embodiments that are within the spirit and scope of the invention as defined by the appended claims.
The following examples illustrate the invention in greater detail, without restricting it. Further compounds according to the invention, of which the preparation is not explicitly described, can be prepared in an analogous way.
The compounds, which are mentioned in the examples and the salts thereof represent preferred embodiments of the invention as well as a claim covering all subcombinations of the residues of the compound of formula (I) as disclosed by the specific examples.
The term "according to" within the experimental section is used in the sense that the procedure referred to is to be used "analogously to".
EXPERIMENTAL PART
The following table lists the abbreviations used in this paragraph and in the Intermediate Examples and Examples section as far as they are not explained within the text body.
Abbreviation Meaning
br broad
CI chemical ionisation
d doublet
dd doublet of doublet
DAD diode array detector
DCM dichloromethane
DMF N,/V-dimethylform amide
DMSO dimethyl sulfoxide Abbreviation Meaning
ELSD Evaporative Light Scattering Detector
eq. equivalent
ESI electrospray (ES) ionisation
HPLC high performance liquid chromatography
LC-MS liquid chromatography mass spectrometry
m multiplet
MS mass spectrometry
NMR nuclear magnetic resonance spectroscopy : chemical shifts (δ) are given in ppm. The chemical shifts were corrected by setting the DMSO signal to 2.50 ppm using unless otherwise stated.
PDA Photo Diode Array
PoraPak™; a HPLC column obtainable from Waters
q quartet
r.t. or rt room temperature
RT retention time (as measured either with HPLC or UPLC) in minutes s singlet
SM starting material
SQD Single-Quadrupol-Detector
t triplet
THF tetrahydrofuran
UPLC ultra performance liquid chromatography
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.
Specific Experimental Descriptions
NMR peak forms in the following specific experimental descriptions are stated as they appear in the spectra, possible higher order effects have not been considered. Reactions employing microwave irradiation may be run with a Biotage Initator® microwave oven optionally equipped with a robotic unit. The reported reaction times employing microwave heating are intended to be understood as fixed reaction times after reaching the indicated reaction temperature. The compounds and intermediates produced according to the methods of the invention may require purification. Purification of organic compounds is well known to the person skilled in the art and there may be several ways of purifying the same compound. In some cases, no purification may be necessary. In some cases, the compounds may be purified by crystallization. In some cases, impurities may be stirred out using a suitable solvent. In some cases, the compounds may be purified by chromatography, particularly flash column chromatography, using for example prepacked silica gel cartridges, e.g. from Separtis such as Isolute® Flash silica gel or Isolute® Flash NH2 silica gel in combination with a Isolera® autopurifier (Biotage) and eluents such as gradients of e.g. hexane/ethyl acetate or DCM/methanol. In some cases, the compounds may be purified by preparative HPLC using for example a Waters autopurifier equipped with a diode array detector and/or on-line electrospray ionization mass spectrometer in combination with a suitable prepacked reverse phase column and eluents such as gradients of water and acetonitrile which may contain additives such as trifluoroacetic acid, formic acid or aqueous ammonia. In some cases, purification methods as described above can provide those compounds of the present invention which possess a sufficiently basic or acidic functionality in the form of a salt, such as, in the case of a compound of the present invention which is sufficiently basic, a trifluoroacetate or formate salt for example, or, in the case of a compound of the present invention which is sufficiently acidic, an ammonium salt for example. A salt of this type can either be transformed into its free base or free acid form, respectively, by various methods known to the person skilled in the art, or be used as salts in subsequent biological assays. It is to be understood that the specific form (e.g. salt, free base etc) of a compound of the present invention as isolated as described herein is not necessarily the only form in which said compound can be applied to a biological assay in order to quantify the specific biological activity.
The percentage yields reported in the following examples are based on the starting component that was used in the lowest molar amount. Air and moisture sensitive liquids and solutions were transferred via syringe or cannula, and introduced into reaction vessels through rubber septa. Commercial grade reagents and solvents were used without further purification. The term "concentrated in vacuo" refers to use of a Buchi rotary evaporator at a minimum pressure of approximately 15 mm of Hg. All temperatures are reported uncorrected in degrees Celsius ("C).
In order that this invention may be better understood, the following examples are set forth. These examples are for the purpose of illustration only, and are not to be construed as limiting the scope of the invention in any manner. All publications mentioned herein are incorporated by reference in their entirety.
Preparative HPLC conditions
"Purification by preparative HPLC" in the subsequent specific experimental descriptions refers to (unless otherwise noted) the following conditions: Analytics (pre- and post-analytics: Method B):
Waters Aqcuity UPLC-MS: Binary Solvent Manager, Sample
System:
Manager/Organizer, Column Manager, PDA, ELSD, SQD 3001
Column: Aqcuity BEH C18 1 .7 50x2.1 mm
A1 = water + 0.1 % vol. formic acid (99%)
Solvent:
A2 = water + 0.2% vol. ammonia (32%)
B = acetonitrile
Gradient: 0-1 .6 min 1 -99% B, 1 .6-2.0 min 99% B
Flow: 0.8 mL/min
Temperature: 60Ό
Injection: 2.0 μί
Detection: DAD scan range 210-400 nm
MS ESI+, ESI-, scan range 160-1000 m/z
ELSD
Methods: Purify pre.flp
Purify post.flp
Preparation:
Waters Autopurificationsystem: Pump 2545, Sample Manager
System: 2767, CFO,
DAD 2996, ELSD 2424, SQD 3001
Column: XBrigde C18 5μιη 100x30 mm
Solvent: A1 = water + 0.1 % vol. formic acid (99%) A2 = water + 0.2% vol. ammonia (32%)
B = acetonitrile
Gradient: 0-1 min 1 % B, 1 -8 min 1 -99% B, 8-10 min 99% B
Flow: 50 mL/min
Temperature: rt
Solution: max. 250 mg / 2.5 mL dimethyl sufoxide or DMF
Injection: 1 x 2.5 mL
Detection: DAD scan range 210-400 nm
MS ESI+, ESI-, scan range 160-1000 m/z
Flash column chromatography conditions
"Purification by (flash) column chromatography" as stated in the subsequent specific experimental descriptions refers to the use of a Biotage Isolera purification system. For technical specifications see "Biotage product catalogue" on www.biotage.com.
EXAMPLES
Synthetic Intermediates Intermediate 1-1-1
Preparation of [2,6-difluoro-4-(2-methoxyethoxy)phenyl]methanol
Figure imgf000096_0001
25.0 g 3,5-Difluoro-4-(hydroxymethyl)phenol (156 mmol, 1.0 eq.) and 17.6 mL 2- bromoethyl methyl ether (187 mmol, 1 .2 eq.) were dissolved in 1 .2 L DMF. 108 g potassium carbonate (781 mmol, 1 .2 eq.), were added and the reaction mixture was stirred overnight at 60 Ό. The reaction mixture wa s concentrated in vacuoo to half of the volume. The solid was filtered off. The clear solution was concentrated in vacuoo. The residue was dissolved in water and ethyl acetate. The aqueous layer was extracted with ethyl acetate twice. The combined organic layers were dried over silicon filter and concentrated in vacuo. The crude product was used without further purification: 40.8 g, 90% pure, yield 108 %, 168 mmol.
1 H-NMR (300 MHz, DMSO-d6): δ [ppm]= 3.25 (s, 3H), 3.50 - 3.65 (m, 2H), 3.99 - 4.14 (m, 2H), 4.37 (br. s., 2H), 5.07 (br. s., 1 H), 6.52 - 6.75 (m, 2H).
Intermediate 1-2-1
Preparation of 2-(bromomethyl)-1 ,3-difluoro-5- 2-methoxyethoxy)benzene
Figure imgf000097_0001
6.0 g [2,6-Difluoro-4-(2-methoxyethoxy)phenyl]methanol (27.4 mmol, 1 .0 eq.) was dissolved in 22 mL dichloromethane. 1 .9 mL phosphorous tribromide (20.5 mmol, 0.75 eq.) was dissolved in dichloromethane and given to the starting material. The reaction mixture was stirred at room temperature under argon atmosphere for 30 min. The reaction mixture was poured into ice water and the pH was adjusted to 8 by adding saturated sodium hydrogen carbonate solution. It was extracted three times with dichloromethane, the combined organic layers were washed once with water and brine, filtered through a silicone coated filter and dried under reduced pressure to provide the crude product, which was used without further purification: 8.2 g, 19. 2 mmol, 107 % yield.
1 H-NMR (300 MHz, DMSO-d6): δ [ppm]= 3.28 (s, 3H), 3.58 - 3.70 (m, 2H), 4.10 - 4.18 (m, 2H), 4.60 (s, 2H), 6.73 - 6.90 (m, 2H).
Intermediate 1-3-1
Preparation of methyl 1 -[2,6-difluoro-4-(2-methoxyethoxy)benzyl]-1 H-pyrazolo[4,3-b]- pyridine-3-carboxylate
Figure imgf000098_0001
10.0 g Methyl 1 H-pyrazolo[4,3-b]pyridine-3-carboxylate (56.4 mmol, 1 .0 eq. ), CAS Reg. No. 1260891 -66-4, was dissolved in 70 mL DMF and treated with 2.48 g sodiumhydride (60% pure, 62 mmol, 1 .1 eq.). It was stirred at room temperaure under argon atmosphere for one hour.
Then 2.48 g tetrabutyl ammonium iodide (5.64 mmol, 0.1 eq.) was added and the mixture was cooled to 0 Ό. 17.5 g 2-(bromomethyl)- 1 ,3-difluoro-5-(2-methoxy- ethoxy)benzene (62.1 mmol, 1 .1 eq.) dissolved in 66 mL DMF was added dropwise and the reaction mixture was stirred at room temperature over night. The reaction mixture was treated with water and ethyl acetate. It was extracted three times with ethyl acetate, the combined organic layers were washed once with water and brine, filtered through a silicone coated filter and concentrated under reduced pressure. The crude product was purified by flash chromatography. The product fraction was treated with ethyl acetate and strirred for a few minutes. The unsolved precipitate was filtered off, washed with ethyl acetate and dried at 5CC under vacuo. The fi ltrate was dried under reduced pressure. The filtrate was treated with methanol and strirred for a few minutes. The unsolved precipitate was filtered off, washed with methanol. The filtrate was concentrated under reduced pressure. The filtrate and both residues were purified by flash chromatography to provide 6.2 g of the 75% pure target compound: 12.6 mmol,
22%.
1 H-NMR (400 MHz, DMSO-d6): δ [ppm]= 3.28 (s, 3H), 3.58 - 3.66 (m, 2H), 3.89 (s, 3H), 4.09 - 4.16 (m, 2H), 5.76 (s, 2H), 6.77 - 6.86 (m, 2H), 7.57 (dd, 1 H), 8.38 (dd, 1 H), 8.73 (dd, 1 H). The following intermediates were prepared according to the same procedure from commercially available starting materials or from the indicated starting materials (SM):
Figure imgf000099_0001
Intermediate 1-4-1
Preparation of 1 -[2,6-difluoro-4-(2-methoxyethoxy)benzyl]-1 H-pyrazolo[4,3-b]pyridine-3- carboximidamide, salt with hydrochloric acid
Figure imgf000100_0001
1 .96 g Ammonium chloride (36.7 mmol, 5.0 eq.) was suspended in 45 mL toluene and cooled down to 0Ό. Then 18 mL trimethyl aluminium solution (2.0 M in toluene, 37 mmol, 5.0 eq.) was added dropwise. The mixture was allowed to reach 1 CC while adding the trimethyl aluminium solution. It was stirred 10 minutes at 0Ό and 15 minutes at room temperature until no gas was formed any longer. Then 2.77 g methyl 1 -[2,6- difluoro-4-(2-methoxyethoxy)benzyl]-1 H-pyrazolo[4,3-b]pyridine-3-carboxylate 1-3-1 (7.34 mmol, 1 .0 eq.) suspended in 13 mL toluene was added and it was stirred at 80Ό over night. The reaction mixture was cooled down with an ice bath and treated carefully with methanol. It was stirred at room temperature for one hour. The unsoved salts were filtered off and wasched with methanol. The filtrate was concentrated under reduced pressure to provide the target compound, which was used without further purification: 85% pure, 3.1 1 g, 90%, 6.64 mmol.
1 H-NMR (400 MHz, DMSO-d6) : δ [ppm]= 3.28 (s, 3H), 3.58 - 3.66 (m, 2H), 3.89 (s, 3H), 4.09 - 4.16 (m, 2H), 5.76 (s, 2H), 6.77 - 6.86 (m, 2H), 7.57 (dd, 1 H), 8.38 (dd, 1 H), 8.73 (dd, 1 H).
The following intermediates were prepared according to the same procedure from the indicated starting materials (SM) :
Figure imgf000100_0002
Figure imgf000101_0001
Intermediate 1-5-1
Preparation of 2-{1 -[2,6-difluoro-4-(2-methoxyethoxy)benzyl]-1 H-pyrazolo[4,3-b]pyridin- 3-yl}-5-methoxypyrimidin-4-amine
Figure imgf000101_0002
1 .5 g 1 -[2,6-Difluoro-4-(2-methoxyethoxy)benzyl]-1 H-pyrazolo[4,3-b]pyridine-3-carbox- imidamide, salt with hydrochloric acid, 1-4-1 (3.77 mmol, 1 .0 eq.) was suspended in isoamyl alcohol and treated with 646 mg 3,3-bis(dimethylamino)-2- methoxypropanenitrile (3.77 mmol, 1 .0 eq.) and 750 μΙ_ piperidine (750 μιηοΙ, 0.2 eq.). It was stirred at 1 10Ό over night under argon atmosph ere. The reaction mixture was concentrated under reduced pressure and purified by flash chromatography and HPLC to provide the target compound as salt in 100% purity: 260 mg. It was dissolved in methylene chloride and a few drops methanol and treated with saturated sodium hydogen carbonate solution. It was stirred for 5 minutes and the organic layer was filtered through a silicone coated filter and concentrated under reduced pressure: 237 mg, 100% pure, 14 % yield, 0.54 mmol.
1 H-NMR (400 MHz, DMSO-d6): δ [ppm]= 3.27 (s, 3H), 3.57 - 3.65 (m, 2H), 3.88 (s, 3H), 4.07 - 4.15 (m, 2H), 5.67 (s, 2H), 6.70 - 6.98 (m, 4H), 7.46 (dd, 1 H), 7.96 (s, 1 H), 8.24 (d, 1 H), 8.59 (dd, 1 H).
The following intermediates were prepared according to the same procedure from the indicated starting materials (SM):
Figure imgf000102_0001
methoxypyrimi
din-4-amine
Intermediate 1-6-1
Preparation of 1 -(4-ethoxy-2,6-difluorobenzyl)-1 H-pyrazolo[3,4-c]pyridine-3-carbox- imidamide, salt with hydrochloric acid
Figure imgf000103_0001
3.00 g 1 -(4-Ethoxy-2,6-difluorobenzyl)-1 H-pyrazolo[3,4-c]pyridine-3-carbonitrile (9.55 mmol, 1 .0 eq.) 1-7-1 was suspended in 25 mL methanol. 920 μΙ_ sodium methylate solution (5.4 M in methanol, 5.0 mmol, 0.5 eq.) was added and this mixture was stirred overnight at 50 "C. Under heating the reaction mixt ure became a clear, dark solution. 766 mg ammonia chloride (14.3 mmol, 1 .5 eq.) and 220 μΙ_ acetic acid (3.9 mmol, 0.4 eq.) were added. The reaction mixture was diluted with 10 mL methanol. The mixture was stirred at 50 Ό overnight. Under heating the r eaction mixture turned clear. The reaction mixture was concentrated under vacuo. The residue was diluted with dichloromethane and 1 M hydrochloric acid. The layers were separated and the aqueous layer was extracted with dichloromethane twice. The combined organic layers were dried using a waterresistant filter and concentrated under reduced pressure. The aqueous layer was concentrated under reduced pressure to provide the target compound with 90% purity: 4.12 g, 10.1 mmol, 106%.
1 H NMR (400 MHz, DMSO-d6) δ [ppm] = 1 .29 (t, 3H), 4.04 (q, 2H), 5.99 (s, 2H), 6.74 - 6.89 (m, 2H), 8.18 (dd, 1 H), 8.57 (d, 1 H), 9.57 (s, 2H), 9.62 - 9.74 (m, 3H).
Intermediate 1-7-1
Preparation of 1 -(4-ethoxy-2,6-difluorobenzyl)-1 H-pyrazolo[3,4-c]pyridine-3-carbonitrile
Figure imgf000104_0001
6.7 g 3-Bromo-1 -(4-ethoxy-2,6-difluorobenzyl)-1 H-pyrazolo[3,4-c]pyridine 1-3-2 (18.3 mmol, 1 .0 eq.) was dissolved in 65 mL DMF and 2.1 g zinc cynanide (18.3 mmol, 1.0 eq.) and 4.5 g [1 ,1 ' -bis(diphenylphosphino)ferrocene]dichloropalladium(ll), complex with dichloromethane (5.5 mmol, 0.3 eq.) were added. The reaction mixture was stirred at 120 Ό for 3 d. Water and ethyl acetate were add ed. The organic layer was extracted with water three times, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by chromatography: 3.62 g, 80 % pure, 50% yield, 9.2 mmol. 1 H-NMR (300 MHz, DMSO-d6): δ [ppm]= 1 .27 (t, 3H), 4.01 (q, 2H), 5.89 (s, 2H), 6.72 - 6.84 (m, 2H), 7.90 (dd, 1 H), 8.47 (d, 1 H), 9.48 (s, 1 H).
Intermediate 1-8-1
Preparation of methyl 1 ,4,6,7-tetrahydropyrano[4,3-c]pyrazole-3-carboxylate
Figure imgf000104_0002
3.0 g 1 ,4,6,7-Tetrahydropyrano[4,3-c]pyrazole-3-carboxylic acid (17.8 mmol, 1 .0 eq.) was supended in 75 mL methanol. 18 mL hydrochloric acid solution (4 M in dioxane, 71 mmol, 4.0 eq.) was added. The reaction mixture was stirred 1 day at 60 Ό and 3 days at room temperature. The reaction mixture was concentrated in vacuo and suspended in ethyl acetate. The suspension was washed with saturated sodiumbicarbonate solution, with water twice and with bringe. The organic layer waws dried over a silicon filter and concentrated in vacuo to provide the 75% pure crude product which was used without further purification: 1 .6 g, 6.5 mmol, 36%
1 H NMR (400 MHz, DMSO-d6) δ [ppm] = 2.60 - 2.73 (m, 2H), 3.67 - 3.84 (m, 5H), 4.56 - 4.71 (m, 2H), 13.14 - 13.71 (m, 1 H). EXAMPLE COMPOUNDS Example 2-1-1
Preparation of 2-{1 -[2,6-difluoro-4-(2-methoxyethoxy)benzyl]-1 H-pyrazolo[4,3-b]pyridin- 3-yl}-5-methoxy-N-(3-methoxypyridin-4-yl)pyrimidin-4-amine
Figure imgf000105_0001
1 15 mg 2-{1 -[2,6-Difluoro-4-(2-methoxyethoxy)benzyl]-1 H-pyrazolo[4,3-b]pyridin-3-yl}-5- methoxypyrimidin-4-amine 1-5-1 (260 μιηοΙ, 1 .0 eq.), 58.4 mg 4-bromo-3- methoxypyridine hydrochloride (1 :1 ) (260 μιηοΙ, 1 .0 eq.), 254 mg cesium carbonate (780 μιηοΙ, 3.0 eq.), 5.8 mg palladium (II) acetate (26 μιηοΙ, 0.1 eq.) and 22.6 mg 4,5- bis(diphenylphosphino)-9,9-dimethylxanthene (39 μιηοΙ, 0.15 eq.) were combined in 1 mL DMF and stirred at 105Ό over night. The reactio n mixture was treated with dichloromethane and water and stirred for a few minutes. The organic layer was filtered through a silicone coated filter and concentrated under reduced pressure. The crude product was treated with methanol and the unsolved precipitate was filtered off, washed with methanol and dried at 50Ό under vacuo: 88 mg, 100% pure, 62% yield, 0.16 mmol.
1 H-NMR (400 MHz, DMSO-d6): δ [ppm]= 3.26 (s, 3H), 3.54 - 3.64 (m, 2H), 4.03 (s, 3H), 4.07 (s, 3H), 4.09 - 4.16 (m, 2H), 5.74 (s, 2H), 6.76 - 6.90 (m, 2H), 7.55 (dd, 1 H), 8.07 (s, 1 H), 8.26 (d, 1 H), 8.30 - 8.41 (m, 3H), 8.81 (dd, 1 H), 9.63 (d, 1 H).
The following examples were prepared according to the same procedure from the indicated starting materials (SM = starting material):
Figure imgf000106_0001
Figure imgf000107_0001
Example 2-2-1
Preparation of 2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl) pyrimidin-2-yl}-1 H-pyrazolo[4,3-b]pyridin-1 -yl)methyl]phenoxy}ethanol
Figure imgf000108_0001
80 mg 2-{1 -[2,6-Dif luoro-4-(2-methoxyethoxy)benzyl]-1 H-pyrazolo[4,3-b]pyridin-3-yl}-5- methoxy-N-(3-methoxypyridin-4-yl)pyrimidin-4-amine 2-1 -1 (146 μιηοΙ, 1 .0 eq.) was dissolved in 3.7 mL dichloromethane. At 0Ό the 470 μΙ_ boron tribromide solution (1 M in dichloromethane, 470 μιηοΙ, 3.2 eq.) was added drop wise. It was stirred at this temperature for 30 minutes under argon atmosphere. The reaction mixture was quenshed with ice water and the pH was adjusted to 10 by addition of sodium hydroxide solution (2 M). It was extracted 3 times with dichloromethane / isopropanole 9:1 , washed once with water and brine, filtered through a silicone coated filter and concentrated in vacuo. The crude product was purified by HPLC chromatography under basic conditions: 21 mg, 95% pure, 26% yield, 37 μιηοΙ.
1 H-NMR (400 MHz, DMSO-d6): δ [ppm]= 3.67 (q, 2H), 3.96 - 4.10 (m, 8H), 4.90 (br t, 1 H), 5.74 (s, 2H), 6.75 - 6.84 (m, 2H), 7.55 (dd, 1 H), 8.07 (s, 1 H), 8.26 (d, 1 H), 8.30 - 8.40 (m, 3H), 8.81 (dd, 1 H), 9.63 (d, 1 H).
The following example was prepared according to the same procedure from the indicated starting material (SM = starting material):
2-2-2 2-[3,5-difluoro-4- 1 H-NMR (400MHz,
({3-[5-methoxy-4- DMSO-de): δ [ppm]=
SM = H (pyridazin-4- 3.67 (t, 2H), 3.95 - 4.10 2-1 -4 ylamino)pyrimidin (m, 5H), 4.94 (br s, 1 H),
-2-yl]-1 H- 5.74 (s, 2H), 6.75 - 6.86 pyrazolo[4,3- (m, 2H), 7.54 (dd, 1 H), b]pyridin-1 - 8.27 - 8.39 (m, 2H),
Figure imgf000108_0002
yl}methyl)phenox 8.82 (dd, 1 H), 8.97 (br yjethanol d, 1 H), 9.34 (br s, 1 H), 9.58 - 9.85 (m, 2H).
Example 2-3-1
Preparation of 2-[1 -(4-ethoxy-2,6-difluorobenzyl)-5,5-dioxido-1 ,4,6,7-tetrahydrothio- pyrano[4,3-c]pyrazol-3- l]-5-methox -N- ridin-4-yl)pyrimidin-4-amine
Figure imgf000109_0001
50 mg 2-[1 -(4-Ethoxy-2,6-difluorobenzyl)-1 ,4,6,7-tetrahydrothiopyrano[4,3-c]pyrazol-3- yl]-5-methoxy-N-(pyridin-4-yl)pyrimidin-4-amine 2-1-5 (98 μηιοΙ, 1 .0 eq.) was dissolved in chloroform and was cooled to 0 Ό. Then 34 mg 3- chloroperbenzoic acid (196 μιηοΙ, 2.0 eq.) were added and the reaction mixture was stirred for 2 d at rt. Again 34 mg 3- chloroperbenzoic acid (196 μιηοΙ, 2.0 eq.) were added and the reaction mixture was stirred for 3 d at rt. Dichloromethan and sodium thiosulfate-solution (10%) was added. It was stirred for 10 min at rt. After seperating the layers the aqueous layer was extracted with dichloromethan three times. The collected organic layers were extracted with water and brine, dried with silicon filter and concentrated in vacuo. The crude product was purified by HPLC: 9 mg, 99% pure, 17% yield, 0.02 mmol.
1 H-NMR (300 MHz, DMSO-d6): δ [ppm]= 1 .31 (t, 3H), 3.30 - 3.40 (m, 2H), 3.48 - 3.59 (m, 2H), 3.99 (s, 3H), 4.06 (q, 2H), 4.50 (s, 2H), 5.28 (s, 2H), 6.78 - 6.88 (m, 2H), 8.08 (d, 2H), 8.24 (s, 1 H), 8.33 - 8.40 (m, 2H), 8.46 (s, 1 H).
Biological investigations
The following assays can be used to illustrate the commercial utility of the compounds according to the present invention. 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 calculated utilizing data sets obtained from testing of one or more synthetic batch.
Biological Assay 1.0:
Bub1 kinase assay
Bub1 -inhibitory activities of compounds described in the present invention were quantified using a time-resolved fluorescence energy transfer (TR-FRET) kinase assay which measures phosphorylation of the synthetic peptide Biotin-Ahx-VLLPKKSFAEPG (C-terminus in amide form), purchased from e.g. Biosyntan (Berlin, Germany) by the (recombinant) catalytic domain of human Bub1 (amino acids 704-1085), expressed in Hi5 insect cells with an N-terminal His6-tag and purified by affinity- (Ni-NTA) and size exclusion chromatography.
In a typical assay 11 different concentrations of each compound (0.1 nM, 0.33 nM, 1 .1 nM, 3.8 nM, 13 nM, 44 nM, 0.15 μΜ, 0.51 μΜ, 1 .7 μΜ, 5.9 μΜ and 20 μΜ) were tested in duplicate within the same microtiter plate. To this end, 100-fold concentrated compound solutions (in DMSO) were previously prepared by serial dilution (1 :3.4) of 2 mM stocks in a clear low volume 384-well source microtiter plate (Greiner Bio-One, Frickenhausen, Germany), from which 50 nL of compounds were transferred into a black low volume test microtiter plate from the same supplier. Subsequently, 2 μί of Bub1 (the final concentration of Bub1 was adjusted depending on the activity of the enzyme lot in order to be within the linear dynamic range of the assay: typically ~ 200 ng/mL were used) in aqueous assay buffer [50 mM Tris/HCI pH 7.5, 10 mM magnesium chloride (MgCI2), 200 mM potassium chloride (KCI), 1 .0 mM dithiothreitol (DTT), 0.1 mM sodium ortho-vanadate, 1 % (v/v) glycerol, 0.01 % (w/v) bovine serum albumine (BSA), 0.005% (v/v) Trition X-100 (Sigma), 1 x Complete EDTA-free protease inhibitor mixture (Roche)] were added to the compounds in the test plate and the mixture was incubated for 15 min at 22Ό to allow pre-equilibration of th e putative enzyme-inhibitor complexes before the start of the kinase reaction, which was initiated by the addition of 3 μΙ_ 1.67- fold concentrated solution (in assay buffer) of adenosine-tri-phosphate (ATP, 10 μΜ final concentration) and peptide substrate (1 μΜ final concentration). The resulting mixture (5 μΙ_ final volume) was incubated at 22Ό d uring 60 min., and the reaction was stopped by the addition of 5 μΙ_ of an aqueous EDTA-solution (50 mM EDTA, in 100 mM HEPES pH 7.5 and 0.2 % (w/v) bovine serum albumin) which also contained the TR- FRET detection reagents (0.2 μΜ streptavidin-XL665 [Cisbio Bioassays, Codolet, France] and 1 nM anti-phosho-Serine antibody [Merck Millipore, cat. # 35-001 ] and 0.4 nM LANCE EU-W1024 labeled anti-mouse IgG antibody [Perkin-Elmer, product no. AD0077, alternatively a Terbium -cryptate-labeled anti-mouse IgG antibody from Cisbio Bioassays can be used]). The stopped reaction mixture was further incubated 1 h at 22Ό in order to allow the formation of complexes b etween peptides and detection reagents. Subsequently, the amount of product was evaluated by measurement of the resonance energy transfer from the Eu-chelate-antibody complex recognizing the Phosphoserine residue to the streptavidin-XL665 bound to the biotin moiety of the peptide. To this end, the fluorescence emissions at 620 nm and 665 nm after excitation at 330-350 nm were measured in a TR-FRET plate reader, e.g. a Rubystar or Pherastar (both from BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin-Elmer) and the ratio of the emissions (665 nm/622 nm) was taken as indicator for the amount of phosphorylated substrate. The data were normalised using two sets of (typically 32-) control wells for high- (= enzyme reaction without inhibitor = 0 % = Minimum inhibition) and low- (= all assay components without enzyme = 100 % = Maximum inhibition) Bub1 activity. IC5o values were calculated by fitting the normalized inhibition data to a 4- parameter logistic equation (Minimum, Maximum, IC5o, Hill; Y = Max + (Min - Max) / (1 + (X/IC5o)Hill)).
Biological Assay 2.1 :
Proliferation Assay:
Human tumour cells were originally obtained from the American Type Culture Collection (ATCC), the Deutsche Sammlung von Mikroorganismen und Zellkulturen (DSMZ, German Collection of Microorganisms and Cell Cultures), or Epo GmbH Berlin. Cultivated HeLa human cervical tumor cells (DSMZ ACC-57) were plated at a density of 3000 cells/well in a 96-well multititer plate in 200 μΙ_ of growth medium supplemented 10% fetal calf serum. After 24 hours, the cells of one plate (zero-point plate) were stained with crystal violet (see below), while the medium of the other plates was replaced by fresh medium containing the test substances in various concentrations (0 μΜ, as well as in the range of 0.001 -10 μΜ; the final concentration of the solvent dimethyl sulfoxide was 0.5%). The cells were incubated for 4 days in the presence of test substances. Cell proliferation was determined by staining the cells with crystal violet: the cells were fixed by adding 20 μ-, ιτΐθθβιιπΓ ι point of an 1 1 % glutaric aldehyde solution for 15 minutes at room temperature. After three washing cycles of the fixed cells with water, the plates were dried at room temperature. The cells were stained by adding 100 μΙ-ymeasuring point of a 0.1 % crystal violet solution (pH 3.0). After three washing cycles of the stained cells with water, the plates were dried at room temperature. The dye was dissolved by adding 100 μΙ-ymeasuring point of a 10% acetic acid solution. Absorbtion was determined by photometry at a wavelength of 595 nm. The change of cell number, in percent, was calculated by normalization of the measured values to the absorbtion values of the zero-point plate (=0%) and the absorbtion of the untreated (0 μιη) cells (=100%). The IC50 values were determined by means of a 4 parameter fit.
Compounds had been evaluated in the HeLa human cervical cancer cell line to demonstrate antiproliferative activity.
Biological Assay 2.2:
Proliferation Assay:
Human tumour cells were originally obtained from the American Type Culture Collection (ATCC), the Deutsche Sammlung von Mikroorganismen und Zellkulturen (DSMZ, German Collection of Microorganisms and Cell Cultures), or Epo GmbH Berlin. Cultivated HeLa human cervical tumor cells (DSMZ ACC-57) were plated at a density of 3000 cells/well in a 96-well multititer plate in 200 μΙ_ of growth medium supplemented 10% fetal calf serum. After 24 hours, the cells of one plate (zero-point plate) were stained with crystal violet (see below), while the medium of the other plates was supplemented with the test substances in various concentrations (0 μΜ, as well as in the range of 0.001 -10 μΜ; the final concentration of the solvent dimethyl sulfoxide was adjusted to 0.1 %) using a Hewlett-Packard HP D300 Digital Dispenser. The cells were incubated for 4 days in the presence of test substances. Cell proliferation was determined by staining the cells with crystal violet: the cells were fixed by adding 20 μΙ-ymeasuring point of an 11 % glutaric aldehyde solution for 15 minutes at room temperature. After three washing cycles of the fixed cells with water, the plates were dried at room temperature. The cells were stained by adding 100 μυιηθθβιιπης point of a 0.1 % crystal violet solution (pH 3.0). After three washing cycles of the stained cells with water, the plates were dried at room temperature. The dye was dissolved by adding 100 μυιηθθβιιπης point of a 10% acetic acid solution. Absorbtion was determined by photometry at a wavelength of 595 nm. The change of cell number, in percent, was calculated by normalization of the measured values to the absorbtion values of the zero-point plate (=0%) and the absorbtion of the untreated (0 μιη) cells (=100%). The IC50 values were determined by means of a 4 parameter fit.
Compounds had been evaluated in the HeLa human cervical cancer cell line to demonstrate antiproliferative activity.
Biological Assay 3.1 :
Proliferation Assay (HeLa+Paclitaxel):
Cultivated HeLa human cervical tumor cells (DSMZ ACC-57) were plated at a density of 3000 cells/well in a 96-well multititer plate in 100 μΙ_ of growth medium supplemented 10% fetal calf serum. After 24 hours, the cells of one plate (zero-point plate) were stained with crystal violet (see below), while the medium of the other plates was replaced by fresh culture medium (100 μΙ_) containing 3 nM of paclitaxel (Sigma- Aldrich). After 4 hours of incubation at 37Ό, 100 μΙ_ of culture medium containing 3 nM of paclitaxel and containing the test substances in various concentrations (0 μΜ, as well as in the range of 0.002-20 μΜ to achieve final concentrations in the range of 0.001 -10 μΜ; the final concentration of the solvent dimethyl sulfoxide was adjusted to 0.5%) was added. The cells were incubated for another 92 hours at 37Ό in the presence of test substances. Cell proliferation was determined by staining the cells with crystal violet: the cells were fixed by adding 20 μΙ-ymeasuring point of an 1 1 % glutaric aldehyde solution for 15 minutes at room temperature. After three washing cycles of the fixed cells with water, the plates were dried at room temperature. The cells were stained by adding 100 μ-,/ιτΐθθβιιπης point of a 0.1 % crystal violet solution (pH 3.0). After three washing cycles of the stained cells with water, the plates were dried at room temperature. The dye was dissolved by adding 100 μ-, ιτΐθθβιιπΓ ι point of a 10% acetic acid solution. Absorbtion was determined by photometry at a wavelength of 595 nm. The change of cell number, in percent, was calculated by normalization of the measured values to the absorbtion values of the zero-point plate (=0%) and the absorbtion of the untreated (0 μιη) cells (=100%). The IC5o values were determined by means of a 4 parameter fit.
Biological Assay 3.2:
Proliferation Assay (HeLa+Paclitaxel):
Cultivated HeLa human cervical tumor cells (DSMZ ACC-57) were plated at a density of 3000 cells/well in a 96-well multititer plate in 100 μΙ_ of growth medium supplemented 10% fetal calf serum. After 24 hours, the cells of one plate (zero-point plate) were stained with crystal violet (see below), while the medium of the other plates was replaced by fresh culture medium containing 3 nM of paclitaxel (Sigma-Aldrich). After 4 hours of incubation at 37Ό, test substances were a dded in various concentrations (0 μΜ, as well as in the range of 0.001 -10 μΜ; the final concentration of the solvent dimethyl sulfoxide was adjusted to 0.1 %) using a Hewlett-Packard HP D300 Digital Dispenser. The cells were incubated for another 92 hours at 37Ό in the presence of test substances. Cell proliferation was determined by staining the cells with crystal violet: the cells were fixed by adding 20 μυιηθθβιιπης point of an 1 1 % glutaric aldehyde solution for 15 minutes at room temperature. After three washing cycles of the fixed cells with water, the plates were dried at room temperature. The cells were stained by adding 100 μυιηθθβιιπης point of a 0.1 % crystal violet solution (pH 3.0). After three washing cycles of the stained cells with water, the plates were dried at room temperature. The dye was dissolved by adding 100 μυιηθθβιιπης point of a 10% acetic acid solution. Absorbtion was determined by photometry at a wavelength of 595 nm. The change of cell number, in percent, was calculated by normalization of the measured values to the absorbtion values of the zero-point plate (=0%) and the absorbtion of the untreated (0 μιη) cells (=100%). The IC5o values were determined by means of a 4 parameter fit.
Biological Assay 4.1 : Formation-Assay
Cell-based Mechanistic Assay: Changes of phosphorylation status of histone 2A by inhibition of kinase activity of Bub1
This assay determines the suppression of histone 2A phosphorylation by a Bub1 kinase inhibitor during co-treatment with Nocodazole. 25000 cells (cells were ordered from ATCC) are seeded in 96well plate for 5 h at 37Ό. C ells are treated with Nocodazole (^g/ml) and varying concentrations (between 3nM and 10μΜ) of test compounds for 16h. Cells are fixed (20 min, Fixing solution R&D), washed three times with PBS and blocked with Odyssey blocking buffer before incubating with the primary antibody against phosphorylated H2A (5μς/ιτιΙ ABIN482721 ) overnight at 2-8Ό. After washing, secondary IRDye-labeled antibody mix with cell stains is added for 1 h and washed again with PBS. Plates are scanned with LiCor Odyssey Infrared Imager CLx at 800nm for P-H2A and at 700nm for cell stains Draq5/Sapphire. The quotient of 800nm and 700nm for Nocodazole only treated cells is set as 100% and the quotient of 800nm and 700nm of untreated cells is set as 0%. The results given as % are reflecting the inhibition of Bub1 kinase activity compared to control and normalized according to cell number. The IC5o values are determined by means of a 4 parameter fit.
Biological Assay 4.2: Abrogation-Assay
Cell-based Mechanistic Assay: Changes of phosphorylation status of pre-induced phospho-histone 2A by inhibition of kinase activity of Bub1
This assay measures the inhibition of histone 2A phosphorylation, which was induced by pre-treatment of the cells with Nocodazole, by a Bub1 kinase inhibitor. 25000 cells (cells were ordered from ATCC) are seeded in 96well plate for 5 h at 37Ό. Cells are treated with Nocodazole (^g/ml). After 16h varying concentrations (between 3nM and 10μΜ) of test compounds are added and the cells are incubated for another 1 h. Cells are fixed (20min, Fixing solution R&D), washed three times with PBS and blocked with Odyssey blocking buffer before incubating with the primary antibody against phosphorylated H2A (5Mg/ml ABIN482721 ) overnight at 2-8<C. After washing, secondary IRDye-labeled antibody mix with cell stains is added for 1 h and washed again with PBS. Plates are scanned with LiCor Odyssey Infrared Imager CLx at 800nm for P-H2A and at 700nm for cell stains Draq5/Sapphire. The quotient of 800nm and 700nm for Nocodazole only treated cells is set as 100% and the quotient of 800nm and 700nm of untreated cells is set as 0%. The results given as % are reflecting the inhibition of Bub1 kinase activity compared to control and normalized according to cell number. The IC5o values are determined by means of a 4 parameter fit.
Histone H2A is an immediate intracellular substrate of Bub1 kinase. Determination of the phosphorylation status of Histone H2A provides a direct measure of the intracellular activity of Bub1 kinase.
Biological Assay 5:
Evaluation of drug-drug interaction potential with paclitaxel
To evaluate the drug-drug interaction potential of test compounds with paclitaxel in vivo 8 mg/kg of paclitaxel are injected once intravenously into the tail vein of NMRI nude mice. Immediately thereafter 50 mg/kg of the test compound is administered by gavage to mice. Blood is taken from mice following decapitation 1 , 3, 7 and 24 hours after injection of Paclitaxel. Plasma concentrations of test compound and of paclitaxel, respectively, are determined by LC/MSMS. The data from the paclitaxel mono treatment group, the test compound mono treatment group, and the combination treatment group are compared for evaluation of the drug-drug interaction potential.
The following table gives the data for the examples of the present invention for the biological assaysl , 2.1 , 2.2, 3.1 and 3.2:
Table 1 :
Biological Biological Biological Biological Biological
Assay 1 : Assay 2.1 : Assay 2.2: Assay 3.1 : Assay 3.2:
Example Bub1 kinase Proliferation Proliferation Proliferation Proliferation
No. assay assay (HeLa assay (HeLa assay (HeLa assay (HeLa mean ICso cell line) cell line) + paclitaxel) + paclitaxel)
[mol/L] ICso [mol/L] ICso [mol/L] ICso [mol/L] ICso [mol/L]
2-1 -1 4.2 E-8 nd nd nd nd
2-1 -2 5.0 E-8 nd 1.6 E-6 nd 7.4 E-7
2-1 -3 2.1 E-7 nd nd nd nd
2-1 -4 3.2 E-6 nd nd nd nd
2-1 -5 9.8 E-9 3.0 E-6 nd 4.5 E-7 nd
2-1 -6 3.8 E-8 3.8 E-6 nd 9.3 E-7 nd
2-2-1 5.8 E-8 nd 2.4 E-6 nd 4.9 E-7
2-2-2 4.3 E-6 nd >1.0 E-5 nd >1.0 E-5
2-3-1 5.4 E-6 nd nd nd nd

Claims

Claims
1 A compound of formula I)
Figure imgf000117_0001
in which
V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH or CR5, or,
V and Y represent N, and W and Z, independently of each other, represent CH or CR5, R1 and R2 represent, independently of each other, a hydrogen atom or a halogen atom,
R3 represents a hydrogen atom or a group selected from:
hydroxy, Ci -C6-alkyl, Ci -C6-haloalkyl, Ci -C6-hydroxyalkyl,
(Ci -C3-alkoxy)-(Ci -C6-alkyl)-, C3-C6-cycloalkyl, (C3-C6-cycloalkyl)-(Ci -C3-alkyl)-,
Ci -Ce-alkoxy, Ci -Ce-haloalkoxy, (C2-C6-hydroxyalkyl)-0-, (Ci -C3-alkoxy)-(C2-C6- alkoxy)-, C3-C6-cycloalkoxy and (C3-C6-cycloalkyl)-(Ci -C3-alkoxy)-,
wherein said hydroxyalkyl groups are optionally substituted with one, two or three halogen atoms selected from:
fluorine and chlorine, represents a hydrogen atom, or a group selected from: Ci-Ce-alkyl, C3-C6-cycloalkyl, Ci-C6-haloalkyl, C2-C6-hydroxyalkyl and
(Ci-C3-alkoxy)-(C2-C6-alkyl)-,
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci -C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy,
(C2-C3-hydroxyalkyl)-0-, (Ci -C3-alkoxy)-(C2-C3-alkoxy)-, Ci-C3-haloalkoxy, -N(H)C(=0)H,
Figure imgf000118_0001
-N(H)C(=0)-(C3-C4-cycloalkyl), -N(H)C(=0)-(Ci-C3-hydroxyalkyl), -C(=0)N(R9)2, -N(H)C(=0)N(H)R9 and -C(=0)OR10,
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from:
*-X-(CH2)2-#,
Figure imgf000118_0002
*-(CH2)2-X-,
*-X-(CH2)3-#,
Figure imgf000118_0003
*-(CH2)3-X-#,
*-C(R11)=N-C(R11)=C(R11)-#,
and
*-C(R11)=C(R11)-C(R11)=N-#,
in which groups X represents -NH-, -N(Ci-C3-alkyl)-, -0-, -S-, -S(=0)- or
-S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule,
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl, Ci -C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -C3-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-,
R10 represents a hydrogen atom or a group selected from: Ci -C3-alkyl, Ci -C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl,
(C3-C4-cycloalkyl)-(Ci -C3-alkyl)- and (Ci -C3-alkoxy)-(C2-C3-alkyl)-,
R1 1 represents, independently of each other, a hydrogen atom or a halogen atom or a group selected from:
methyl, ethyl and Ci -C2-haloalkyl, or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
2. The compound according to claim 1 ,
wherein:
V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH or CR5,
R1 and R2 represent, independently of each other, a hydrogen atom or a halogen atom, represents a hydrogen atom or a group selected from:
Ci -Ce-alkoxy, Ci -C6-haloalkoxy, (C2-C6-hydroxyalkyl)-0-, (Ci -C3-alkoxy)-(C2-C6- alkoxy)-, C3-C6-cycloalkoxy and (C3-C6-cycloalkyl)-(Ci -C3-alkoxy)-,
R4 represents a hydrogen atom, or a group selected from:
Ci -Ce-alkyl, C3-C6-cycloalkyl, Ci -Ce-haloalkyl, C2-C6-hydroxyalkyl and
(Ci -C3-alkoxy)-(C2-C6-alkyl)-,
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci -C3-alkyl, C3-C4-cycloalkyl, Ci -C3-haloalkyl, Ci -C3-alkoxy, Ci -C3- haloalkoxy and -C(=0)N(R9)2, R7 and R8 are linked to one another in such a way that they jointly form a group selected from:
*-X-(CH2)3-#,
Figure imgf000120_0001
*-(CH2)3-X-#,
*-C(R11)=N-C(R11)=C(R11)-#,
and
*-C(R11)=C(R11)-C(R11)=N-#,
in which groups X represents -NH-, -N(Ci-C3-alkyl)-, -0-, -S-, -S(=0)- or
-S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule,
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl, C2-C3-hydroxyalkyl, C3-C4-cycloalkyl and (Ci-C3-alkoxy)-(C2-C3- alkyl)-,
R11 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group, or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
3. The compound according to claim 1 or 2,
wherein:
V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH or CR5, R1 and R2 represent, independently of each other, a hydrogen atom or a halogen atom,
R3 represents a hydrogen atom or a group selected from:
Ci-C3-alkoxy, (C2-C3-hydroxyalkyl)-0- and (Ci-C3-alkoxy)-(C2-C3-alkoxy)-,
R4 represents a hydrogen atom, or a group selected from:
Ci-C3-alkyl and C2-C3-hydroxyalkyl,
R5 represents, independently of each other, a halogen atom or a group selected from:
cyano, Ci-C3-alkyl, Ci-C3-alkoxy and -C(=0)N(R9)2,
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from:
Figure imgf000121_0001
*-C(R11)=N-C(R11)=C(R11)-#,
and
*-C(R11)=C(R11)-C(R11)=N-#,
in which groups X represents -0-, -S-, or -S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule,
R9 represents, independently of each other, a hydrogen atom or a group selected from:
Ci-C3-alkyl and C3-C4-cycloalkyl,
R11 represents, independently of each other, a hydrogen atom or a halogen atom or a methyl group, or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
4. The compound according to any of claims 1 to 3,
wherein:
V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH or CR5,
R1 and R2 represent, independently of each other, a halogen atom,
R3 represents a group selected from:
Ci-C3-alkoxy, (C2-C3-hydroxyalkyl)-0- and (Ci-C3-alkoxy)-(C2-C3-alkoxy)-,
R4 represents a Ci-C3-alkyl group,
R5 represents, independently of each other, a group selected from:
Ci-C3-alkyl and Ci-C3-alkoxy,
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from:
Figure imgf000122_0001
*-CH=N-CH=CH-#,
and
*-CH=CH-CH=N-#,
in which groups X represents -0-, -S-, or -S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule, or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
5. The compound according to any of claims 1 to 4,
wherein:
V, W, Y and Z, independently of each other, represent CH or CR5,
or,
V represents N, and W, Y and Z, independently of each other, represent CH or CR5, or,
W represents N, and V, Y and Z, independently of each other, represent CH,
R1 and R2 represent a fluorine atom,
R3 represents a group selected from:
ethoxy, 2-hydroxyethoxy and 2-methoxyethoxy,
R4 represents a methyl group,
R5 represents, independently of each other, a group selected from:
methyl and methoxy,
R7 and R8 are linked to one another in such a way that they jointly form a group
selected from:
Figure imgf000123_0001
*-CH=N-CH=CH-#,
and
*-CH=CH-CH=N-#,
in which groups X represents -0-, -S-, or -S(=0)2-,
and,
in which * represents the point of attachment of R7 to the rest of the molecule, and in which # represents the point of attachment of R8 to the rest of the molecule, or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
6. A compound of formula (I) according to any of claims 1 to 5, which is selected from the group consisting of:
2-{1 -[2,6-difluoro-4-(2-methoxyethoxy)benzyl]-1 H-pyrazolo[4,3-b]pyridin-3-yl}-5- methoxy-N-(3-methoxypyridin-4-yl)pyrimidin-4-amine,
2-[1 -(4-ethoxy-2,6-difluorobenzyl)-1 H-pyrazolo[3,4-c]pyridin-3-yl]-5-methoxy-N-(3- methoxypyridin-4-yl)pyrimidin-4-amine,
2-[1 -(4-ethoxy-2,6-difluorobenzyl)-1 H-pyrazolo[3,4-c]pyridin-3-yl]-5-methoxy-N-(2- methylpyrimidin-4-yl)pyrimidin-4-amine,
N-(2-{1 -[2,6-difluoro-4-(2-methoxyethoxy)benzyl]-1 H-pyrazolo[4,3-b]pyridin-3-yl}-5- methoxypyrimidin-4-yl)pyridazin-4-amine,
2-[1 -(4-ethoxy-2,6-difluorobenzyl)-1 ,4,6,7-tetrahydrothiopyrano[4,3-c]pyrazol-3-yl]-5- methoxy-N-(pyridin-4-yl)pyrimidin-4-amine,
2-[1 -(4-ethoxy-2,6-difluorobenzyl)-1 ,4,6,7-tetrahydropyrano[4,3-c]pyrazol-3-yl]-5- methoxy-N-(pyridin-4-yl)pyrimidin-4-amine,
2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl)amino]pyrimidin-2-yl}-1 H- pyrazolo[4,3-b]pyridin-1 -yl)methyl]phenoxy}ethanol,
2-[3,5-difluoro-4-({3-[5-methoxy-4-(pyridazin-4-ylamino)pyrimidin-2-yl]-1 H-pyrazolo- [4,3-b]pyridin-1 -yl}methyl)phenoxy]ethanol, and
2-[1 -(4-ethoxy-2,6-difluorobenzyl)-5,5-dioxido-1 ,4,6,7-tetrahydrothiopyrano[4,3-c]- pyrazol-3-yl]-5-methoxy-N-(pyridin-4-yl)pyrimidin-4-amine, or an N-oxide, a salt, a tautomer or a stereoisomer of said compound, or a salt of said N-oxide, tautomer or stereoisomer.
7. Use of a compound of general formula (I) according to any of claims 1 to 6 for the treatment or prophylaxis of a disease.
8. Use of a compound of general formula (I) according to claim 7, whereby the disease is hyperproliferative disease and/or a disorder responsive to induction of cell death.
9. Use of a compound of general formula (I) according to claim 8, whereby the hyperproliferative disease and/or disorder responsive to induction of cell death is a haematological tumour, solid tumour and/or metastases thereof.
10. Use of a compound of general formula (I) according to according to claim 9, whereby the hyperproliferative disease is cervical cancer.
1 1. A pharmaceutical composition comprising at least one compound of general formula 5 (I) according to any of claims 1 to 6, together with at least one pharmaceutically
acceptable carrier or auxiliary.
12. A composition according to claim 1 1 for the treatment of a haematological tumour, a solid tumour and/or metastases thereof.
o
13. A combination comprising one or more first active ingredients selected from a compound of general formula (I) according to any of claims 1 to 6, and one or more second active ingredients selected from chemotherapeutic anti-cancer agents and target-specific anti-cancer agents.
5
PCT/EP2017/056076 2016-03-18 2017-03-15 Annulated pyrazoles as bub1 kinase inhibitors for treating proliferative disorders WO2017157992A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP16161045 2016-03-18
EP16161045.6 2016-03-18

Publications (1)

Publication Number Publication Date
WO2017157992A1 true WO2017157992A1 (en) 2017-09-21

Family

ID=55586204

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2017/056076 WO2017157992A1 (en) 2016-03-18 2017-03-15 Annulated pyrazoles as bub1 kinase inhibitors for treating proliferative disorders

Country Status (1)

Country Link
WO (1) WO2017157992A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108546266A (en) * 2018-07-25 2018-09-18 上海毕得医药科技有限公司 A kind of synthetic method of 1,4,6,7- oxinanes [4,3-C] pyrazoles -3- carboxylic acids
US10266548B2 (en) 2011-10-06 2019-04-23 Bayer Intellectual Property Gmbh Substituted benzylindazoles for use as Bub1 kinase inhibitors in the treatment of hyperproliferative diseases
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10350206B2 (en) 2014-09-19 2019-07-16 Bayer Pharma Aktiengesellschaft Benzyl substituted indazoles as BUB1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5011472A (en) 1988-09-06 1991-04-30 Brown University Research Foundation Implantable delivery system for biological factors
WO2012112363A1 (en) 2011-02-14 2012-08-23 Merck Sharp & Dohme Corp. Cathepsin cysteine protease inhibitors
WO2013050438A1 (en) 2011-10-06 2013-04-11 Bayer Pharma Aktiengesellschaft Substituted benzylindazoles for use as bub1 kinase inhibitors in the treatment of hyperproliferative diseases.
WO2013092512A1 (en) 2011-12-21 2013-06-27 Bayer Intellectual Property Gmbh Substituted benzylpyrazoles
WO2013167698A1 (en) 2012-05-11 2013-11-14 Bayer Pharma Aktiengesellschaft Substituted cycloalkenopyrazoles as bub1 inhibitors for the treatment of cancer
WO2014147204A1 (en) 2013-03-21 2014-09-25 Bayer Pharma Aktiengesellschaft Heteroaryl substituted indazoles
WO2014147203A1 (en) 2013-03-21 2014-09-25 Bayer Pharma Aktiengesellschaft 3-heteroaryl substituted indazoles
WO2014202583A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Substituted benzylpyrazoles
WO2014202588A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Heteroaryl substituted pyrazoles
WO2014202590A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Substituted benzylpyrazoles
WO2014202584A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Heteroaryl substituted pyrazoles
WO2015063003A1 (en) 2013-10-30 2015-05-07 Bayer Pharma Aktiengesellschaft Heteroaryl substituted pyrazoles

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5011472A (en) 1988-09-06 1991-04-30 Brown University Research Foundation Implantable delivery system for biological factors
WO2012112363A1 (en) 2011-02-14 2012-08-23 Merck Sharp & Dohme Corp. Cathepsin cysteine protease inhibitors
WO2013050438A1 (en) 2011-10-06 2013-04-11 Bayer Pharma Aktiengesellschaft Substituted benzylindazoles for use as bub1 kinase inhibitors in the treatment of hyperproliferative diseases.
WO2013092512A1 (en) 2011-12-21 2013-06-27 Bayer Intellectual Property Gmbh Substituted benzylpyrazoles
WO2013167698A1 (en) 2012-05-11 2013-11-14 Bayer Pharma Aktiengesellschaft Substituted cycloalkenopyrazoles as bub1 inhibitors for the treatment of cancer
WO2014147204A1 (en) 2013-03-21 2014-09-25 Bayer Pharma Aktiengesellschaft Heteroaryl substituted indazoles
WO2014147203A1 (en) 2013-03-21 2014-09-25 Bayer Pharma Aktiengesellschaft 3-heteroaryl substituted indazoles
WO2014202583A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Substituted benzylpyrazoles
WO2014202588A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Heteroaryl substituted pyrazoles
WO2014202590A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Substituted benzylpyrazoles
WO2014202584A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Heteroaryl substituted pyrazoles
WO2015063003A1 (en) 2013-10-30 2015-05-07 Bayer Pharma Aktiengesellschaft Heteroaryl substituted pyrazoles

Non-Patent Citations (39)

* Cited by examiner, † Cited by third party
Title
"Isotopic Compositions of the Elements", PURE APPL. CHEM., vol. 70, no. 1, 1997, pages 217 - 235
A. E. MUTLIB ET AL., TOXICOL. APPL. PHARMACOL., vol. 169, 2000, pages 102
A. M. SHARMA ET AL., CHEM. RES. TOXICOL., vol. 26, 2013, pages 410
AIELLO ET AL., NEW ENGL. J. MED., vol. 331, 1994, pages 1480
B. TESTA ET AL., INT. J. PHARM., vol. 19, no. 3, 1984, pages 271
BARON A ET AL., ELIFE, vol. 5, 2016, pages E12187
BOLANOS-GARCIA VM; BLUNDELL TL, TRENDS BIOCHEM. SCI., vol. 36, 2010, pages 141
C. J. WENTHUR ET AL., J. MED. CHEM., vol. 56, 2013, pages 5208
C. L. PERRIN ET AL., J. AM. CHEM. SOC., vol. 127, 2005, pages 9641
C. L. PERRIN ET AL., J. AM. CHEM. SOC., vol. 129, 2007, pages 4490
ELOWE S, MOL. CELL. BIOL., vol. 31, 2011, pages 3085
F. MALTAIS E, J. MED. CHEM., vol. 52, 2009, pages 7993
F. SCHNEIDER ET AL., ARZNEIM. FORSCH. / DRUG. RES., vol. 56, 2006, pages 295
GOODMAN; GILMAN'S ET AL.: "The Pharmacological Basis of Therapeutics", 1996, MCGRAW-HILL, pages: 1225 - 1287
HANAHAN D; WEINBERG RA, CELL, vol. 100, 2000, pages 57
HANAHAN D; WEINBERG RA, CELL, vol. 144, 2011, pages 646
KAWASHIMA ET AL., SCIENCE, vol. 327, 2010, pages 172
KING RW, BIOCHIM BIOPHYS ACTA, vol. 1786, 2008, pages 4
KOPS GJ ET AL., NATURE REV. CANCER, vol. 5, 2005, pages 773
LOPEZ ET AL., INVEST. OPTHTHALMOL. VIS. SCI., vol. 37, 1996, pages 855
MUSACCHIO A; SALMON ED, NAT. REV. MOL. CELL. BIOL, vol. 8, 2007, pages 379
NEMA, S. ET AL.: "Excipients and Their Use in Injectable Products", PDA JOURNAL OF PHARMACEUTICAL SCIENCE & TECHNOLOGY, vol. 51, no. 4, 1997, pages 166 - 171
P. KOCIENSKI: "Protecting Groups", 2000, THIEME MEDICAL PUBLISHERS
PEER ET AL., LAB. INVEST., vol. 72, 1995, pages 638
POWELL, M.F. ET AL.: "Compendium of Excipients for Parenteral Formulations", PDA JOURNAL OF PHARMACEUTICAL SCIENCE & TECHNOLOGY, vol. 52, no. 5, 1998, pages 238 - 311, XP009119027
PURE APPL CHEM, vol. 45, 1976, pages 11 - 30
RICKE ET AL., J. CELL BIOL., vol. 199, 2012, pages 931 - 949
RIEDER CL; MAIATO H, DEV. CELL, vol. 7, 2004, pages 637
ROBERTS BT ET AL., MOL. CELL BIOL., vol. 14, 1994, pages 8282
S. M. BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
SCHMIDT M; BASTIANS H, DRUG RES. UPDATES, vol. 10, 2007, pages 162
SCHMIDT M; MEDEMA RH, CELL CYCLE, vol. 5, 2006, pages 159
STRICKLEY, R.G: "Parenteral Formulations of Small Molecule Therapeutics Marketed in the United States (1999)-Part-1", PDA JOURNAL OF PHARMACEUTICAL SCIENCE & TECHNOLOGY, vol. 53, no. 6, 1999, pages 324 - 349
SUIJKERBUIJK SJ; KOPS GJ, BIOCHEM. BIOPHYS. ACTA, vol. 1786, 2008, pages 24
T. W. GREENE: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
T.W. GREENE; P.G.M. WUTS: "Protective Groups in Organic Synthesis", 1999, WILEY
WATANABE Y, COLD SPRING HARB. SYMP. QUANT. BIOL., vol. 75, 2010, pages 419
WEAVER BA; CLEVELAND DW, CANCER RES., vol. 67, 2007, pages 10103
YUAN B ET AL., CLIN. CANCER RES., vol. 12, 2006, pages 405

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10266548B2 (en) 2011-10-06 2019-04-23 Bayer Intellectual Property Gmbh Substituted benzylindazoles for use as Bub1 kinase inhibitors in the treatment of hyperproliferative diseases
US10604532B2 (en) 2011-10-06 2020-03-31 Bayer Intellectual Property Gmbh Substituted benzylindazoles for use as BUB1 kinase inhibitors in the treatment of hyperproliferative diseases
US10350206B2 (en) 2014-09-19 2019-07-16 Bayer Pharma Aktiengesellschaft Benzyl substituted indazoles as BUB1 inhibitors
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
CN108546266A (en) * 2018-07-25 2018-09-18 上海毕得医药科技有限公司 A kind of synthetic method of 1,4,6,7- oxinanes [4,3-C] pyrazoles -3- carboxylic acids

Similar Documents

Publication Publication Date Title
AU2016212230B2 (en) 4H-pyrrolo[3,2-c]pyridin-4-one derivatives
JP6664385B2 (en) Benzyl-substituted indazoles as Bub1 inhibitors
EP3700904B1 (en) 4h-pyrrolo[3,2-c]pyridin-4-one derivatives
EP3331611B1 (en) 1h-pyrrol-3-amines
EP3010901B1 (en) Heteroaryl substituted pyrazoles
WO2016042080A1 (en) Benzyl substituted indazoles as bub1 kinase inhibitors
EP2976336A1 (en) 3-heteroaryl substituted indazoles
JP2017529353A (en) Benzyl-substituted indazoles as Bub1 kinase inhibitors
EP3063138A1 (en) Heteroaryl substituted pyrazoles
JP2017530963A (en) Benzyl-substituted indazoles
WO2017157992A1 (en) Annulated pyrazoles as bub1 kinase inhibitors for treating proliferative disorders
WO2017148995A1 (en) 1-(pyrimidin-2-yl)-1h-indazoles having bub1 kinase inhibiting activity
WO2017157991A1 (en) 1-alkyl-pyrazoles and -indazoles as bub1 inhibitors for the treatment of hyperproliferative diseases
WO2018104307A1 (en) Aromatic sulfonamide derivatives and their use as anatagon i sts or negative allosteric modulators of p2x4
EP4244225A1 (en) N-[2-({4-[3-(anilino)-4-oxo-4,5,6,7-tetrahydro-1h-pyrrolo[3,2-c]pyridin-2-yl]pyridin-3-yl)oxy)ethyl]prop-2-enamide derivatives and similar compounds as egfr inhibitors for the treatment of cancer

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 17710301

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 17710301

Country of ref document: EP

Kind code of ref document: A1