Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMBINATION OF BUB1 INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2018/158175
Kind Code:
A1
Abstract:
The present invention relates to combinations of at least two components, component A and component B, component A being a Bub1-inhibitor of general formula (I), or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a pharmaceutically acceptable salt, or a mixture of same, and component B being an anti-hyperproliferative, cytotoxic and/or cytostatic agent selected from a taxane, such as docetaxel or paclitaxel, or combinations thereof.

Inventors:
SIEMEISTER GERHARD (DE)
BONE WILHELM (DE)
MENGEL ANNE (DE)
Application Number:
PCT/EP2018/054630
Publication Date:
September 07, 2018
Filing Date:
February 26, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BAYER PHARMA AG (DE)
International Classes:
A61K31/506; A61P35/00
Domestic Patent References:
WO2016042084A12016-03-24
WO2016042084A12016-03-24
WO2013050438A12013-04-11
WO2013092512A12013-06-27
WO2013167698A12013-11-14
WO2014147144A12014-09-25
WO2014147203A12014-09-25
WO2014147204A12014-09-25
WO2014202586A12014-12-24
WO2014202590A12014-12-24
WO2014202588A12014-12-24
WO2014202584A12014-12-24
WO2015063003A12015-05-07
WO2014202583A12014-12-24
WO2015193339A12015-12-23
WO2016041925A12016-03-24
WO2016042080A12016-03-24
WO2016042081A12016-03-24
WO2016120196A12016-08-04
WO2016202755A12016-12-22
WO2017021348A12017-02-09
WO2017102649A12017-06-22
Foreign References:
EP15200590A2015-12-16
US5023252A1991-06-11
US5011472A1991-04-30
Other References:
BARON ANNA P ET AL: "Probing the catalytic functions of Bub1 kinase using the small molecule inhibitors BAY-320 and BAY-524", ELIFE, vol. 5, 17 February 2016 (2016-02-17), XP002780498
HITCHCOCK MARION ET AL: "Abstract 2718: Synthesis and characterization of novel benzylpyrazole-based BUB1 kinase inhibitors with anti-tumor activity", CANCER RESEARCH, vol. 76, no. Suppl. 14, July 2016 (2016-07-01), & 107TH ANNUAL MEETING OF THE AMERICAN-ASSOCIATION-FOR-CANCER-RESEARCH (AACR); NEW ORLEANS, LA, USA; APRIL 16 -20, 2016, XP002780499, DOI: 10.1158/1538-7445.AM2016-2718
ANONYMOUS: "Paclitaxel", 13 April 2016 (2016-04-13), XP002780500, Retrieved from the Internet [retrieved on 20180425]
HANAHAN D; WEINBERG RA, CELL, vol. 100, 2000, pages 57
HANAHAN D; WEINBERG RA, CELL, vol. 144, 2011, pages 646
MUSACCHIO A, CURR. BIOL., vol. 25, 2015, pages R1002 - R1018
KAWASHIMA ET AL., SCIENCE, vol. 327, 2010, pages 172
WATANABE Y, COLD SPRING HARB. SYMP. QUANT. BIOL., vol. 75, 2010, pages 419
BARON ET AL., ELIFE, vol. 5, 2016, pages e12187
RICKE ET AL., J. CELL BIOL., vol. 199, 2012, pages 931
PURE APPL CHEM, vol. 45, 1976, pages 11 - 30
S. M. BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
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
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
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
MOUNTAIN CF; LUNG: "Revisions in the International System for Staging Lung Cancer", CHEST, vol. 111, no. 6, 1997, pages 1710 - 7
EDGE SB, BYRD DR, COMPTON CC, ET AL.: "AJCC Cancer Staging Manual, 7th ed.", 2010, SPRINGER, article LUNG, pages: 253 - 70
"Merck Index, 11th ed.", 1996, .
MOLINOFF ET AL.: "Goodman and Gilman's The Pharmacological Basis of Therapeutics, 9th ed.", 1996, MCGRAW-HILL, pages: 1225 - 1287
CHOU T.C., PHARMACOL. REV., vol. 58, 2006, pages 621
Attorney, Agent or Firm:
BIP PATENTS (DE)
Download PDF:
Claims:
CLAIMS

1 . A combination of at least two components, component A and component B, comprising:

- a component A being an inhibitor of Bub1 which is, 2-{3,5-difluoro-4-[(3-{5-methoxy-4- [(3-methoxypyridin-4-yl)amino]pyrimidin-2-yl}-1 /-/-indazol-1 -yl)methyl]phenoxy}ethanol, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a pharmaceutically acceptable salt, or a mixture of same; and,

- a component B being an anti-hyperproliferative, cytotoxic and/or cytostatic agent selected from a taxane, such as docetaxel or paclitaxel, or combinations thereof, for use in the treatment or prophylaxis of a disorder selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC), Non-Small Cell Lung Cancer (NSCLC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof.

2. A combination of at least two components, component A and component B, comprising a component A being an inhibitor of Bub1 of general formula (I),

V, W, Y and Z independently of each other represent CH or CR2, wherein one of V, W,

V and Z represents CR2'

or,

V represents N, and W, Y and Z independently of each other represent CH or CR2, or,

V and Y represent N, and W and Z independently of each other represent CH or CR2, R1 represents a group selected from :

Ci -Ce-alkyl, Ci-C6-haloalkyl, C3-C6-cycloalkyl, (Ci-C3-alkoxy)-(C2-C3-alkyl)-, and (C3-C6-cycloalkyl)-(Ci-C3-alkyl)-,

R2 represents, independently of each other, halogen or a group selected from: Ci -C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci -C3-alkoxy,

Ci -C3-haloalkoxy, -N(H)C(=0)-(Ci-C3-alkyl), -N(H)C(=0)H, -N(H)C(=0)-(Ci-C3-hydroxyalkyl),

-N(H)C(=0)-(Ci-C3-alkyl)-(Ci-C3-alkoxy), -N(H)C(=0)-phenyl, -N(H)C(=0)-(C3-C4-cycloalkyl),

-N(H)C(=0)-(Ci-C3-alkyl)-(C3-C4-cycloalkyl), and -N(H)C(=0)N(H)R8,

said -N(H)C(=0)-phenyl being optionally substituted at the phenyl ring, one, two or three times, identically or differently, with a substituent selected from:

halogen, hydroxy, cyano, Ci-C4-alkyl, Ci-C4-haloalkyl,

Ci -C4-alkoxy, Ci -C4-haloalkoxy, C3-C4-cycloalkyl, and

C3-C -cycloalkyloxy,

said -N(H)C(=0)-(C3-C -cycloalkyl) being optionally substituted at the C3-C -cycloalkyl ring with a substituent selected from:

fluorine, chlorine, trifluoromethyl, and methoxy,

R3 represents a group selected from :

C2-C6-hydroxyalkyl, and R4,

said C2-C6-hydroxyalkyl groups being optionally substituted with one, two or three halogen atoms selected from:

fluorine, and chlorine,

R4 represents -(C2-C6-alkyl)-OC(=0)-C(H)(R5)-N(H)C(=0)-C(H)(R7)-NH2, in which C2-C6-alkyl is optionally substituted with one, two or three halogen atoms selected from: fluorine, and chlorine,

R5 and R7 independently of each other represent hydrogen (glycine) or a group selected from:

-CH3 (alanine), -C(H)(CH3)2 (valine), -(CH2)2CH3 (norvaline), -CH2C(H)(CH3)2 (leucine), - C(H)(CH3)CH2CH3 (isoleucine), -(CH2)3CH3 (norleucine), -C(CH3)3 (2-fert-butylglycine), benzyl (phenylalanine), 4-hydroxybenzyl (tyrosine), -(CH2)3NH2 (ornithine), -(CH2)4NH2 (lysine), -(CH2)2C(H)(OH)CH2NH2 (hydroxylysine), -CH2OH (serine), -(CH2)2OH (homoserine), -C(H)(OH)CH3 (threonine), -(CH2)3N(H)C(=NH)NH2 (arginine), - (CH2)3N(H)C(=0)NH2 (citrulline), -CH2C(=0)NH2 (asparagine), -CH2C(=0)OH (aspartic acid), -(CH2)2C(=0)OH (glutamic acid), -(CH2)2C(=0)NH2 (glutamine), -CH2SH (cysteine), -(CH2)2SH (homocysteine), -(CH2)2SCH3 (methionine), -CH2SCH3 (S- methylcysteine), (1 /-/-imidazol-4-yl)methyl- (histidine), (1 /-/-indol-3-yl)methyl- (thryptophan), -CH2NH2 (2,3-diaminopropanoic acid), and -(CH2)2NH2 (2,4- diaminobutanoic acid),

R8 represents hydrogen 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)-, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a pharmaceutically acceptable salt, or a mixture of same,

and component B being an anti-hyperproliferative, cytotoxic and/or cytostatic agent selected from a taxane, such as docetaxel or paclitaxel, or combinations thereof, for use in the treatment or prophylaxis of a disorder selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC), Non-Small Cell Lung Cancer (NSCLC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof.

3. The combination for use according to claim 2, wherein,

V, W, Y and Z independently of each other represent CH or CR2, wherein one of V, W, Y and Z represents CR2 or,

V represents N, and W, Y and Z independently of each other represent CH or CR2,

R1 represents a group selected from :

Ci-C3-alkyl, Ci-C3-haloalkyl, C3-C4-cycloalkyl,

(Ci-C3-alkoxy)-(C2-C3-alkyl)-, and (C3-C4-cycloalkyl)-(Ci-C3-alkyl)-,

R2 represents, independently of each other, halogen or a group selected from: Ci-C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy, Ci-C3-haloalkoxy, -N(H)C(=0)-(Ci-C3-alkyl), -N(H)C(=0)H,

-N(H)C(=0)-(Ci-C3-hydroxyalkyl),

-N(H)C(=0)-(Ci-C3-alkyl)-(Ci-C3-alkoxy), -N(H)C(=0)-phenyl, -N(H)C(=0)-(C3-C4-cycloalkyl),

-N(H)C(=0)-(Ci-C3-alkyl)-(C3-C4-cycloalkyl), and -N(H)C(=0)N(H)R8,

said -N(H)C(=0)-phenyl being optionally substituted at the phenyl ring, one, two or three times, identically or differently, with a substituent selected from:

halogen, hydroxy, cyano, Ci-C4-alkyl, Ci-C4-haloalkyl,

Ci-C -alkoxy, Ci-C -haloalkoxy, C3-C -cycloalkyl, and

C3-C -cycloalkyloxy,

said -N(H)C(=0)-(C3-C -cycloalkyl) being optionally substituted at the C3-C -cycloalkyl ring with a substituent selected from:

fluorine, chlorine, trifluoromethyl, and methoxy,

R3 represents a group selected from :

C2-C6-hydroxyalkyl, and R4,

said C2-C6-hydroxyalkyl groups being optionally substituted with one, two or three halogen atoms selected from:

fluorine, and chlorine, R4 represents -(C2-C6-alkyl)-OC(=0)-C(H)(R5)-N(H)C(=0)-C(H)(R7)-NH2, in which C2-C6-alkyl is optionally substituted with one, two or three halogen atoms selected from:

fluorine, and chlorine, R5 and R7 independently of each other represent hydrogen (glycine) or a group selected from: -CH3 (alanine), -C(H)(CH3)2 (valine), -(CH2)2CH3 (norvaline), -CH2C(H)(CH3)2 (leucine), - C(H)(CH3)CH2CH3 (isoleucine), -(CH2)3CH3 (norleucine), -C(CH3)3 (2-fert-butylglycine), benzyl (phenylalanine), 4-hydroxybenzyl (tyrosine), -(CH2)3NH2 (ornithine), -(CH2)4NH2 (lysine), -(CH2)2C(H)(OH)CH2NH2 (hydroxylysine), -CH2OH (serine), -(CH2)2OH (homoserine), -C(H)(OH)CH3 (threonine), -(CH2)3N(H)C(=NH)NH2 (arginine), - (CH2)3N(H)C(=0)NH2 (citrulline), -CH2C(=0)NH2 (asparagine), -CH2C(=0)OH (aspartic acid), -(CH2)2C(=0)OH (glutamic acid), -(CH2)2C(=0)NH2 (glutamine), -CH2SH (cysteine), -(CH2)2SH (homocysteine), -(CH2)2SCH3 (methionine), -CH2SCH3 (S- methylcysteine), (1 /-/-imidazol-4-yl)methyl- (histidine), (1 /-/-indol-3-yl)methyl- (thryptophan), -CH2NH2 (2,3-diaminopropanoic acid), and -(CH2)2NH2 (2,4- diaminobutanoic acid),

R8 represents hydrogen 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)-, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a pharmaceutically acceptable salt, or a mixture of same.

4. The combination for use according to claim 2 or 3, wherein,

V, W, Y and Z independently of each other represent CH or CR2, wherein one of V, W,

Y and Z represents CR2

or,

V represents N, and W, Y and Z independently of each other represent CH or CR2,

R1 represents a group selected from :

Ci-C3-alkyl, Ci-C3-haloalkyl, and C3-C4-cycloalkyl, R2 represents, independently of each other, halogen or a group selected from: Ci-C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy,

Ci-C3-haloalkoxy, -N(H)C(=0)H,

-N(H)C(=0)-(Ci-C3-hydroxyalkyl),

-N(H)C(=0)-(Ci-C3-alkyl)-(Ci-C3-alkoxy), -N(H)C(=0)-phenyl, -N(H)C(=0)-(C3-C4-cycloalkyl),

-N(H)C(=0)-(Ci-C3-alkyl)-(C3-C4-cycloalkyl), and -N(H)C(=0)N(H)R8, said -N(H)C(=0)-phenyl being optionally substituted at the phenyl ring, one, two or three times, identically or differently, with a substituent selected from:

halogen, hydroxy, cyano, Ci-C4-alkyl, Ci-C4-haloalkyl,

Ci-C4-alkoxy, Ci-C4-haloalkoxy, C3-C -cycloalkyl, and C3-C -cycloalkyloxy,

said -N(H)C(=0)-(C3-C -cycloalkyl) being optionally substituted at the C3-C -cycloalkyl ring with a substituent selected from:

fluorine, chlorine, trifluoromethyl, and methoxy, R3 represents a group selected from :

C2-C6-hydroxyalkyl, and R4,

said C2-C6-hydroxyalkyl group being optionally substituted with one, two or three halogen atoms selected from:

fluorine, and chlorine,

R4 represents -(C2-C6-alkyl)-OC(=0)-C(H)(R5)-N(H)C(=0)-C(H)(R7)-NH2, in which C2-C6-alkyl is optionally substituted with one, two or three halogen atoms selected from:

fluorine, and chlorine,

R5 and R7 independently of each other represent a group selected from:

-CH3 (alanine), -C(H)(CH3)2 (valine), -(CH2)2CH3 (norvaline), -(CH2)3NH2 (ornithine), -

(CH2)4NH2 (lysine), and -(CH2)3N(H)C(=NH)NH2 (arginine), R8 represents hydrogen or a group selected from:

Ci-C3-alkyl, Ci-C3-haloalkyl, C2-C3-hydroxyalkyl, C3-C -cycloalkyl,

(C3-C -cycloalkyl)-(Ci-C3-alkyl)-, and (Ci-C3-alkoxy)-(C2-C3-alkyl)-, 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 combination for use according to any one of claims 2 to 4, wherein, V, W, Y and Z independently of each other represent CH or CR2, wherein one of V, W, Y and Z represents CR2 or,

V represents N, and W, Y and Z independently of each other represent CH or CR2, R1 represents a Ci-C3-alkyl group,

R2 represents, independently of each other, halogen or a group selected from: Ci-C3-alkyl, C3-C4-cycloalkyl, Ci-C3-haloalkyl, Ci-C3-alkoxy,

R3 represents a group selected from :

C2-C6-hydroxyalkyl, and R4,

R4 represents -(C2-C6-alkyl)-OC(=0)-C(H)(R5)-N(H)C(=0)-C(H)(R7)-NH2, R5 and R7 independently of each other represent a group selected from :

-CH3 (alanine), and -(CH2)4NH2 (lysine), 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. The combination for use according to any one of claims 2 to 5, wherein,

V, W, Y and Z independently of each other represent CH or CR2, wherein one of V, W, Y and Z represents CR2

or,

V represents N, and W, Y and Z independently of each other represent CH or CR2, R1 represents a methyl group,

R2 represents, independently of each other, fluorine, chlorine or a group selected from:

methyl, cyclopropyl, difluoromethyl, methoxy, and -N(H)C(=0)-CH3,

R3 represents a group selected from :

-(CH2)2OH, and R4, represents -(CH2)2-OC(=0)-C(H)(R5)-N(H)C(=0)-C(H)(R7)-NH2,

R5 represents -CH3 (alanine),

R7 represents -(CH2)4NH2 (lysine), 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. The combination for use according to any one of claims 2 to 6, wherein,

V, W, Y and Z independently of each other represent CH or CR2, wherein one of V, W, Y and Z represents CR2

or,

V represents N, and W, Y and Z independently of each other represent CH or CR2, R1 represents a methyl group,

R2 represents, independently of each other, fluorine, chlorine or a group selected from:

methyl, cyclopropyl, difluoromethyl, methoxy, -N(H)C(=0)-CH3, -N(H)C(=0)-cyclopropyl, and -N(H)C(=0)N(H)-cyclopropyl,

R3 represents a -(CH2)2OH 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.

8. The combination for use according to any one of claims 2 to 7, wherein the component A is a compound selected from the group consisting of :

2-{4-[(3-{4-[(3-chloropyridin-4-yl)amino]-5-methoxypyrimidin-2-yl}-1 /-/-indazol-1 - yl)methyl]-3,5-difluorophenoxy}ethanol ,

2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(2-methylpyridin-4-yl)amino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethanol ,

2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl)amino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethanol ,

N-{4-[(2-{1 -[2,6-difluoro-4-(2-hydroxyethoxy)benzyl]-1 /-/-indazol-3-yl}-5- methoxypyrimidin-4-yl)amino]pyridin-2-yl}acetamide ,

2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methylpyridin-4-yl)amino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethanol ,

2-{3,5-difluoro-4-[(3-{4-[(3-fluoropyridin-4-yl)amino]-5-methoxypyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethanol ,

2-(4-{[3-(4-{[2-(difluoromethyl)pyridin-4-yl]amino}-5-methoxypyrimidin-2-yl)-1 /-/-indazol- 1 -yl]methyl}-3,5-dif luorophenoxy)ethanol ,

2-{4-[(3-{4-[(2,5-dimethylpyridin-4-yl)amino]-5-methoxypyrimidin-2-yl}-1 /-/-indazol-1 - yl)methyl]-3,5-difluorophenoxy}ethanol ,

2-{4-[(3-{4-[(3-cyclopropylpyridin-4-yl)amino]-5-methoxypyrimidin-2-yl}-1 /-/-indazol-1 - yl)methyl]-3,5-difluorophenoxy}ethanol ,

2-{1 -[2,6-difluoro-4-(2-hydroxyethoxy)benzyl]-1 /-/-indazol-3-yl}-4-[(2-methylpyrimidin-4- yl)amino]pyrimidin-5-ol

2-{3,5-difluoro-4-[(3-{4-[(2-fluoropyridin-4-yl)amino]-5-methoxypyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethanol ,

2-{3,5-difluoro-4-[(3-{4-[(5-fluoro-2-methylpyridin-4-yl)amino]-5-methoxypyrimidin-2-yl}- 1 H-indazol-1 -yl)methyl]phenoxy}ethanol ,

2-[3,5-difluoro-4-({3-[5-methoxy-4-(pyrimidin-4-ylamino)pyrimidin-2-yl]-1 H-indazol-1 - yl}methyl)phenoxy]ethanol

/V-{4-[(2-{1 -[2,6-difluoro-4-(2-hydroxyethoxy)benzyl]-1 H-indazol-3-yl}-5- methoxypyrimidin-4-yl)amino]-5-methylpyridin-2-yl}acetamide ,

2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(2-methylpyridin-4-yl)amino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl L-lysyl-L-alaninate, salt with trifluoroacetic acid , 2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(2-methylpyridin-4-yl)amino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl L-lysyl-L-alaninate ,

2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl)amino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl L-lysyl-L-alaninate, salt with trifluoroacetic acid , 2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl)amino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl L-lysyl-L-alaninate,

N-{4-[(2-{1 -[2,6-difluoro-4-(2-hydroxyethoxy)benzyl]-1 H-indazol-3-yl}-5-methoxy- pyrimidin-4-yl)amino]pyridin-2-yl}cyclopropanecarboxamide ,

1 -cyclopropyl-3-{4-[(2-{1 -[2,6-difluoro-4-(2-hydroxyethoxy)benzyl]-1 H-indazol-3-yl}-5- methoxypyrimidin-4-yl)amino]pyridin-2-yl}urea ,

2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxy-2-methylpyridin-4-yl)amino]pyrimi^ yl}-1 H-indazol-1 -yl)methyl]phenoxy}ethanol ,

2- {3,5-difluoro-4-[(3-{5-methoxy-4-[(5-methoxy-2-m

yl}-1 H-indazol-1 -yl)methyl]phenoxy}ethanol ,

3- {4-[(3-{4-[(2,5-dimethylpyridin-4-yl)amino]-5-methoxypyrimidin-2-yl}-1 H-indazol-1 - yl)methyl]-3,5-difluorophenoxy}propan-1 -ol ,

3-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl)amino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}propan-1 -ol ,

3-{3,5-difluoro-4-[(3-{5-methoxy-4-[(2-methylpyrimidin-4-yl)amino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}propan-1 -ol ,

(2R)-3-{3,5-difluoro-4-[(3-{5-methoxy-4-[(2-methylpyrimidin-4-yl)amino]pyrimidin-2-yl}- 1 H-indazol-1 -yl)methyl]phenoxy}-2-methylpropan-1 -ol ,

(2R)-3-{4-[(3-{4-[(2,5-dimethylpyridin-4-yl)amino]-5-methoxypyrimidin-2-yl}-1 H-indazol- 1 -yl)methyl]-3,5-difluorophenoxy}-2-methylpropan-1 -ol ,

N-[4-({2-[1 -(2,6-difluoro-4-{[(2R)-3-hydroxy-2-methylpropyl]oxy}benzyl)-1 H-indazol-3- yl]-5-methoxypyrimidin-4-yl}amino)pyridin-2-yl]acetamide ,

(2R)-3-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl)amino]pyrimidin-2-yl}- 1 H-indazol-1 -yl)methyl]phenoxy}-2-methylpropan-1 -ol ,

3-{3,5-difluoro-4-[(3-{5-methoxy-4-[(2-methylpyridin-4-yl)amino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}propan-1 -ol ,

2-{4-[(3-{4-[(2,6-dimethylpyrimidin-4-yl)amino]-5-methoxypyrimidin-2-yl}-1 H-indazol-1 - yl)methyl]-3,5-difluorophenoxy}ethanol ,

2-{3,5-difluoro-4-[(3-{4-[(2-fluoropyridin-4-yl)amino]-5-methoxypyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl D-lysyl-L-alaninate, salt with trifluoroacetic acid , 2-{3,5-difluoro-4-[(3-{4-[(2-fluoropyridin-4-yl)amino]-5-methoxypyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl D-lysyl-L-alaninate ,

2-{3,5-difluoro-4-[(3-{4-[(2-fluoropyridin-4-yl)amino]-5-methoxypyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl L-lysyl-L-alaninate, salt with trifluoroacetic acid , and 2-{3,5-difluoro-4-[(3-{4-[(2-fluoropyridin-4-yl)amino]-5-methoxypyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl L-lysyl-L-alaninate, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a pharmaceutically acceptable salt, or a mixture of same.

9. The combination for use according to any one of claims 2 to 8, wherein the component A is 2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4- yl)amino]pyrimidin-2-yl}-1 /-/-indazol-1 -yl)methyl]phenoxy}ethanol,

or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a pharmaceutically acceptable salt, or a mixture of same.

10. The combination for use according to any of claims 1 to 9, wherein component B is paclitaxel. 1 1. The combination for use according to any of claims 1 to 9, wherein component B is docetaxel.

12. The combination for use according to any one of claims 1 to 10, wherein paclitaxel is administered once every 3 weeks.

13. The combination for use according to claim 12, wherein 100 - 175 mg/m2 paclitaxel are administered.

14. The combination for use according to any one of claims 1 to 10, wherein paclitaxel is administered once weekly.

15. The combination for use according to claim 14, wherein 75-90 mg/m2 paclitaxel are administered.

16. The combination for use according to any one of claims 1 to 9 or 1 1 , wherein docetaxel is administered once every 3 weeks.

17. The combination for use according to claim 16, wherein 60 - 100 mg/m2 docetaxel are administered.

18. The combination for use according to any one of claims 1 to 9 or 1 1 , wherein docetaxel is administered once weekly. 19. The combination for use according to claim 18, wherein 30-40 mg/m2 docetaxel are administered.

20. The combination according to any of claims 1 to 19, wherein component A or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, is administered one or more times per day or less than once a day.

21. The combination according to any of claims 1 to 20, wherein from about 0.001 mg/kg to about 200 mg/kg body weight of component A or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof are administered, preferably from about 0.01 mg/kg to about 30 mg/kg body weight of component A are administered.

22. The combination for use according to any one of claims 1 to 21 , wherein the cancer is selected from Triple-negative breast cancer (TNBC), Castration- Resistant Prostate Cancer (CRPC), Non-Small Cell Lung Cancer (NSCLC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof.

23. Use of a combination as defined in any one of claims 1 to 22, for the preparation of a medicament for the treatment or prophylaxis of a disorder selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC), Non-Small Cell Lung Cancer (NSCLC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof.

24. A method of treatment or prophylaxis of a disorder selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC), Non-Small Cell Lung Cancer (NSCLC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof, in a subject, comprising administering to said subject a therapeutically effective amount of a combination as defined in any one of claims 1 to 22.

25. A kit comprising a combination as defined in any one of claims 1 to 22, and, optionally, one or more further pharmaceutical agents C; in which optionally both or either of said compound of general formula (I) and anti-hyperproliferative, cytotoxic and/or cytostatic agent are in the form of a pharmaceutical formulation which is ready for use to be administered simultaneously, concurrently, separately or sequentially, for use in the treatment or prophylaxis of a disorder selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC), Non-Small Cell Lung Cancer (NSCLC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof.

26. A composition containing a combination according to any of claims 1 to 22 together with a pharmaceutically acceptable ingredients.

27. A combination of at least two components, component A and component B, wherein component A and component B are as defined in any one of claims 1 to 22, for use in the treatment or prophylaxis of a cancer selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC), Non-Small Cell Lung Cancer (NSCLC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof, wherein said cancer is resistant and/or insensitive to treatment with standard of care drugs selected from a taxane, such as docetaxel or paclitaxel.

28. A combination of at least two components, component A and component B, wherein component A and component B are as defined in any one of claims 1 to 22, for use in a method to sensitize cancer cells selected from breast cancer, such as Triple- negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC), Non-Small Cell Lung Cancer (NSCLC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof, to paclitaxel or docetaxel.

Description:
Combination of Bub1 Inhibitors

The present invention relates to combinations of at least two components, component A and component B, component A being a Bub1 inhibitor of general formula (I) as described herein, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a pharmaceutically acceptable salt, or a mixture of same, and component B being an anti-hyperproliferative, cytotoxic and/or cytostatic agent selected from a taxane, such as docetaxel or paclitaxel, or combinations thereof, for use in the treatment or prophylaxis of a disease, particularly for the treatment of cancer, more particularly for the treatment of breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor- resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC), Non-Small Cell Lung Cancer (NSCLC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof.

Another aspect of the present invention relates to the use of such combinations as described herein for the preparation of a medicament for the treatment or prophylaxis of a disease, particularly for the treatment of cancer, more particularly for the treatment of breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Non-Small Cell Lung Cancer (NSCLC), Castration- Resistant Prostate Cancer (CRPC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof.

Yet another aspect of the present invention relates to methods of treatment or prophylaxis of a disorder selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC), Non-Small Cell Lung Cancer (NSCLC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof, in a subject, comprising administering to said subject a therapeutically effective amount of a combination as described herein.

Further, the present invention relates to a kit comprising a combination of:

- one or more components A, as defined herein, or a pharmaceutically acceptable salt, solvate, hydrate or stereoisomer thereof ;

- a component B, as defined supra; and optionally

- one or more pharmaceutical agents C;

in which optionally either or both of said components A and B are in the form of a pharmaceutical formulation which is ready for use to be administered simultaneously, concurrently, separately or sequentially, , for use in the treatment or prophylaxis of a disorder selected from breast cancer, Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC), Non-Small Cell Lung Cancer (NSCLC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof.

Another aspect of the present invention relates to a combination as defined herein, for use in the treatment or prophylaxis of a cancer selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC), Non-Small Cell Lung Cancer (NSCLC) melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof, wherein said cancer is resistant and/or insensitive to treatment with standard of care drugs selected from a taxane, such as docetaxel or paclitaxel. Another aspect of the present invention relates to a combination as defined herein, for use in a method to sensitize cancer cells selected from breast cancer, such as Triple- negative breast cancer (TNBC), antiestrogen resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Non-Small Cell Lung Cancer (NSCLC), Castration- Resistant Prostate Cancer (CRPC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof, to paclitaxel or docetaxel.

BACKGROUND Cell cycle deregulation represents one of the classical hallmarks of cancer [Hanahan D and Weinberg RA, Cell 100, 57, 2000; Hanahan D and Weinberg RA, Cell 144, 646, 201 1 ] and consequently cell cycle arrest is the predominant mode of action of a lot of the cancer drugs on the market including ' anti-mitotics ' such as taxanes and vinca alkaloids which stablize respectively depolymerize tubuline and thus compromize the function of the mitotic spindle. The concept of cell cycle checkpoint regulation offers a novel approach to cancer treatment: inactivation of cell cycle checkpoints is considered to drive tumor cells into cell divison despite DNA damage or unattached/misattached chromosomes resulting in a lethal degree of DNA damage or aneuploidy.

The spindle assembly checkpoint (SAC, also known as spindle checkpoint or mitotic checkpoint) controls the accurate attachment of mircrotubules of the spindle device to the kinetochors (the attachment site for microtubules) of the duplicated chromosomes. The SAC 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 [for recent review see Musacchio A, Curr. Biol. 25, R1002-R1018, 2015].

The SAC signal is initiated by multipolar-spindle 1 (Mpsl )-mediated phosphorlyation of Met-Glu-Leu-Thr (MELT) motifs on the kinetochore scaffold 1 (KNL-1 ) protein to generate docking sites for budding uninhibited by benzimidazole 1 (Bub1 )/Bub3 dimers. The N-terminal non-catalytic part of Bub1 represents a scaffold for the assembly of mitotic arrest deficient 1 (MAD1 )/MAD2, BubR1 and centromere protein E (CENP-E) proteins. The inactive open from of MAD2 (o-MAD2) gets converted to the active closed conformation (c-MAD2) which binds cell-division cycle 20 homolog (CDC20) and BubFM to form the diffusible mitotic checkpoint complex (MCC) which inhibits the ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C) and subsequently leads to mitotic arrest. Note that MCC generation requires Bub1 protein, however, it ' s kinase function is not involved.

Bub1 kinase phosphorylates histone H2A at Thr120 within the centromeric region of the duplicated chromosomes thereby creating binding sites for shugoshin proteins (Sgo) 1 and 2, which protect centromeric cohesin from premature degradation, and for the chromosomal passenger complex consisting of INCENP, survivin, borealin and Aurora B (AurB) [Kawashima et al. Science 327, 172, 2010; Watanabe Y, Cold Spring Harb. Symp. Quant. Biol. 75, 419, 2010]. Specific inhibition of Bub1 kinase activity results in reduced levels of Sgo1 and Sgo2 at mitotic centromeres. Furthermore, AurB fails to concentrate at the centromeres and was instead found to be spread over the chromosome arms [Baron et al. eLife 5, e12187, 2016]. AurB activity localized at the centromeric region is essential for the resolution of microtubule - kinetochore attachment errors such as syntelic and merotelic attachments. Dyslocatization of AurB due to Bub1 kinase inactivation strongly compromizes the cells ability to resolve attachment errors and results in an increased rate of chromosome aligment defects [Ricke et al. J. Cell Biol. 199, 931 , 2012], in particular in presence of attachment error inducing agents such as microtubule stabilizers paclitaxel or docetaxel.

However, the state of the art neither discloses the combinations of the present invention comprising an inhibitor of Bub1 kinase and a taxane, such as docetaxel or paclitaxel, or combinations thereof. SUMMARY of the INVENTION

Surprisingly it was observed that by administering a Bub1 kinase of general formula (I) as described herein, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a pharmaceutically acceptable salt, or a mixture of same, in combination with an anti-hyperproliferative, cytotoxic and/or cytostatic agent selected from a taxane, such as docetaxel or paclitaxel, or combinations thereof, a significant effect on tumor growth inhibition was obtained over the respective monotherapies, particularly the combinations of the present invention have shown surprising therapeutic advantages in breast cancer, such as Triple-negative breast cancer (TNBC), prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC) and Non-Small Cell Lung Cancer (NSCLC), melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, bladder cancer, such as bladder transitional cell carcinoma cancer models. Such advantages include improved anti-tumor efficacy of the combination of the present invention over the respective monotherapies, particularly:

- when standard of care drugs provide partial and/or incomplete anti-tumor response, and/or

- when tumor progression/regrowth is not stopped or not delayed due to (acquired or intrinsic) resistance of the tumor to standard of care drugs, and/or

- by sensitizing cancer cells to low and/or lower doses of paclitaxel or docetaxel.

Therefore, in accordance with a first aspect, the present invention provides combinations of at least two components, component A and component B, component of general formula (I),

in which,

V, W, Y and Z independently of each other represent CH or CR 2 , wherein one of V, W, Y and Z represents CR 2 '

or,

V represents N, and W, Y and Z independently of each other represent CH or CR 2 , or,

V and Y represent N, and W and Z independently of each other represent CH or CR 2 ,

R 1 represents a group selected from : Ci -Ce-alkyl, Ci-C 6 -haloalkyl, C3-C 6 -cycloalkyl,

(Ci-C3-alkoxy)-(C2-C 3 -alkyl)-, and (C3-C6-cycloalkyl)-(Ci-C 3 -alkyl)-,

R 2 represents, independently of each other, halogen or a group selected from: Ci -C 3 -alkyl, C3-C 4 -cycloalkyl, Ci-C 3 -haloalkyl, Ci -C 3 -alkoxy,

Ci -C 3 -haloalkoxy, -N(H)C(=0)-(Ci-C 3 -alkyl), -N(H)C(=0)H,

-N(H)C(=0)-(Ci-C 3 -hydroxyalkyl),

-N(H)C(=0)-(Ci-C3-alkyl)-(Ci-C 3 -alkoxy), -N(H)C(=0)-phenyl, -N(H)C(=0)-(C 3 -C 4 -cycloalkyl),

-N(H)C(=0)-(Ci-C3-alkyl)-(C 3 -C 4 -cycloalkyl), and -N(H)C(=0)N(H)R 8 ,

said -N(H)C(=0)-phenyl being optionally substituted at the phenyl ring, one, two or three times, identically or differently, with a substituent selected from:

halogen, hydroxy, cyano, Ci-C 4 -alkyl, Ci-C -haloalkyl,

Ci -C -alkoxy, Ci -C -haloalkoxy, C 3 -C -cycloalkyl, and C3-C 4 -cycloalkyloxy,

said -N(H)C(=0)-(C 3 -C -cycloalkyl) being optionally substituted at the C 3 -C -cycloalkyl ring with a substituent selected from:

fluorine, chlorine, trifluoromethyl, and methoxy, R 3 represents a group selected from:

C2-C6-hydroxyalkyl, and R 4 ,

said C 2 -C 6 -hydroxyalkyl groups being optionally substituted with one, two or three halogen atoms selected from:

fluorine, and chlorine,

R 4 represents -(C 2 -C 6 -alkyl)-OC(=0)-C(H)(R 5 )-N(H)C(=0)-C(H)(R 7 )-NH 2 , in which C2-C6-alkyl is optionally substituted with one, two or three halogen atoms selected from:

fluorine, and chlorine,

R 5 and R 7 independently of each other represent hydrogen (glycine) or a group selected from:

-CH 3 (alanine), -C(H)(CH 3 ) 2 (valine), -(CH 2 ) 2 CH 3 (norvaline), -CH 2 C(H)(CH 3 ) 2 (leucine), - C(H)(CH 3 )CH 2 CH 3 (isoleucine), -(CH 2 ) 3 CH 3 (norleucine), -C(CH 3 ) 3 (2-fert-butylglycine), benzyl (phenylalanine), 4-hydroxybenzyl (tyrosine), -(CH 2 ) 3 NH 2 (ornithine), -(CH 2 ) 4 NH 2 (lysine), -(CH 2 ) 2 C(H)(OH)CH 2 NH 2 (hydroxylysine), -CH 2 OH (serine), -(CH 2 ) 2 OH (homoserine), -C(H)(OH)CH 3 (threonine), -(CH 2 ) 3 N(H)C(=NH)NH 2 (arginine), - (CH 2 ) 3 N(H)C(=0)NH2 (citrulline), -CH 2 C(=0)NH 2 (asparagine), -CH 2 C(=0)OH (aspartic acid), -(CH 2 ) 2 C(=0)OH (glutamic acid), -(CH 2 ) 2 C(=0)NH 2 (glutamine), -CH 2 SH (cysteine), -(CH 2 ) 2 SH (homocysteine), -(CH 2 ) 2 SCH 3 (methionine), -CH 2 SCH 3 (S- methylcysteine), (1 /-/-imidazol-4-yl)methyl- (histidine), (1 /-/-indol-3-yl)methyl- (thryptophan), -CH 2 NH 2 (2,3-diaminopropanoic acid), and -(CH 2 ) 2 NH 2 (2,4- diaminobutanoic acid),

R 8 represents hydrogen or a group selected from:

Ci-C 3 -alkyl, Ci-C 3 -haloalkyl, C 2 -C 3 -hydroxyalkyl, C 3 -C 4 -cycloalkyl,

(C 3 -C 4 -cycloalkyl)-(Ci-C 3 -alkyl)-, and (Ci-C 3 -alkoxy)-(C 2 -C 3 -alkyl)-, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a pharmaceutically acceptable salt, or a mixture of same,

and component B being an anti-hyperproliferative, cytotoxic and/or cytostatic agent selected from a taxane, such as docetaxel or paclitaxel, or combinations thereof, for use in the treatment or prophylaxis of a disorder selected from Triple-negative breast cancer (TNBC), Castration- Resistant Prostate Cancer (CRPC) and Non-Small Cell Lung Cancer (NSCLC) and/or metastases thereof.

The present invention also provides combinations of at least two components, component A and component B as described herein, for use in the treatment or prophylaxis of a disorder selected from melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof.

In accordance with another aspect, the present invention covers uses of a combination as defined herein, for the preparation of a medicament for the treatment or prophylaxis of a disorder selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC) and Non-Small Cell Lung Cancer (NSCLC) and/or metastases thereof. The present invention also covers uses of a combination as defined herein, for the preparation of a medicament for the treatment or prophylaxis of a disorder selected from melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof.

In accordance with another aspect, the present invention covers methods of treatment or prophylaxis of a disorder selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC) and Non-Small Cell Lung Cancer (NSCLC) and/or metastases thereof, in a subject, comprising administering to said subject a therapeutically effective amount of a combination as defined herein.

The present invention also covers methods of treatment or prophylaxis of a disorder selected from melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof, in a subject, comprising administering to said subject a therapeutically effective amount of a combination as defined herein. In accordance with a further aspect, the present invention covers kits comprising a combination as defined herein, and, optionally, one or more further pharmaceutical agents C; in which optionally both or either of said compound of general formula (I) and anti-hyperproliferative, cytotoxic and/or cytostatic agent are in the form of a pharmaceutical formulation which is ready for use to be administered simultaneously, concurrently, separately or sequentially, for use in the treatment or prophylaxis of a disorder selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC) and Non-Small Cell Lung Cancer (NSCLC) and/or metastases thereof. The present invention also covers kits comprising a combination as defined herein, and, optionally, one or more further pharmaceutical agents C; in which optionally both or either of said compound of general formula (I) and anti-hyperproliferative, cytotoxic and/or cytostatic agent are in the form of a pharmaceutical formulation which is ready for use to be administered simultaneously, concurrently, separately or sequentially, for use in the treatment or prophylaxis of a disorder selected from melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof.

In accordance with a further aspect, the present invention covers compositions containing a combination as defined herein, for use in the treatment or prophylaxis of a disorder selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC) and Non-Small Cell Lung Cancer (NSCLC) and/or metastases thereof, together with a pharmaceutically acceptable ingredients. The present invention also covers compositions containing a combination as defined herein, for use in the treatment or prophylaxis of a disorder selected from melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof, together with a pharmaceutically acceptable ingredients.

In accordance with a further aspect, the present invention covers combinations of at least two components, component A and component B, wherein component A and component B are as defined herein, for use in the treatment or prophylaxis of a cancer selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC) and Non-Small Cell Lung Cancer (NSCLC) and/or metastases thereof, wherein said cancer is resistant and/or insensitive to treatment with standard of care drugs selected from a taxane, such as docetaxel or paclitaxel. The present invention also covers combinations of at least two components, component A and component B, wherein component A and component B are as defined herein, for use in the treatment or prophylaxis of a cancer selected from melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof, together with a pharmaceutically acceptable ingredients., wherein said cancer is resistant and/or insensitive to treatment with standard of care drugs selected from a taxane, such as docetaxel or paclitaxel.

In accordance with a further aspect, the present invention covers, combinations of at least two components, component A and component B, wherein component A and component B are as defined herein, for use in a method to sensitize cancer cells selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Non-Small Cell Lung Cancer (NSCLC) and Castration- Resistant Prostate Cancer (CRPC) and/or metastases thereof, to paclitaxel or docetaxel.

The present invention covers, combinations of at least two components, component A and component B, wherein component A and component B are as defined herein, for use in a method to sensitize cancer cells selected from melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof, to paclitaxel or docetaxel.

In accordance to an embodiment of the first aspect, component A is a Bub1 kinase inhibitor of formula (I) supra, wherein,

V, W, Y and Z independently of each other represent CH or CR 2 , wherein one of V, W,

Y and Z represents CR 2

or,

V represents N, and W, Y and Z independently of each other represent CH or CR 2 , R 1 represents a group selected from :

Ci-C 3 -alkyl, Ci-C 3 -haloalkyl, C3-C 4 -cycloalkyl,

(Ci-C 3 -alkoxy)-(C2-C 3 -alkyl)-, and (C 3 -C4-cycloalkyl)-(Ci-C 3 -alkyl)-,

R 2 represents, independently of each other, halogen or a group selected from: Ci-C 3 -alkyl, C 3 -C 4 -cycloalkyl, Ci-C 3 -haloalkyl, Ci-C 3 -alkoxy,

Ci-C 3 -haloalkoxy, -N(H)C(=0)H,

-N(H)C(=0)-(Ci-C 3 -hydroxyalkyl),

-N(H)C(=0)-(Ci-C 3 -alkyl)-(Ci-C 3 -alkoxy), -N(H)C(=0)-phenyl, -N(H)C(=0)-(C 3 -C 4 -cycloalkyl),

-N(H)C(=0)-(Ci-C 3 -alkyl)-(C 3 -C 4 -cycloalkyl), and -N(H)C(=0)N(H)R 8 ,

said -N(H)C(=0)-phenyl being optionally substituted at the phenyl ring, one, two or three times, identically or differently, with a substituent selected from:

halogen, hydroxy, cyano, Ci-C 4 -alkyl, Ci-C 4 -haloalkyl,

Ci-C -alkoxy, Ci-C -haloalkoxy, C 3 -C -cycloalkyl, and

C 3 -C 4 -cycloalkyloxy,

said -N(H)C(=0)-(C 3 -C -cycloalkyl) being optionally substituted at the C 3 -C -cycloalkyl ring with a substituent selected from:

fluorine, chlorine, trifluoromethyl, and methoxy,

R 3 represents a group selected from :

C 2 -C 6 -hydroxyalkyl, and R 4 ,

said C 2 -C 6 -hydroxyalkyl groups being optionally substituted with one, two or three halogen atoms selected from:

fluorine, and chlorine,

R 4 represents -(C 2 -C 6 -alkyl)-OC(=0)-C(H)(R 5 )-N(H)C(=0)-C(H)(R 7 )-NH 2 , in which C 2 -C 6 -alkyl is optionally substituted with one, two or three halogen atoms selected from:

fluorine, and chlorine,

R 5 and R 7 independently of each other represent hydrogen (glycine) or a group selected from:

-CH 3 (alanine), -C(H)(CH 3 ) 2 (valine), -(CH 2 ) 2 CH 3 (norvaline), -CH 2 C(H)(CH 3 ) 2 (leucine), - C(H)(CH 3 )CH 2 CH 3 (isoleucine), -(CH 2 ) 3 CH 3 (norleucine), -C(CH 3 ) 3 (2-fert-butylglycine), benzyl (phenylalanine), 4-hydroxybenzyl (tyrosine), -(CH 2 ) 3 NH 2 (ornithine), -(CH 2 ) 4 NH 2 (lysine), -(CH 2 )2C(H)(OH)CH 2 NH2 (hydroxylysine), -CH 2 OH (serine), -(CH 2 ) 2 OH (homoserine), -C(H)(OH)CH 3 (threonine), -(CH 2 ) 3 N(H)C(=NH)NH 2 (arginine), - (CH 2 ) 3 N(H)C(=0)NH 2 (citrulline), -CH 2 C(=0)NH 2 (asparagine), -CH 2 C(=0)OH (aspartic acid), -(CH 2 ) 2 C(=0)OH (glutamic acid), -(CH 2 ) 2 C(=0)NH 2 (glutamine), -CH 2 SH (cysteine), -(CH 2 ) 2 SH (homocysteine), -(CH 2 ) 2 SCH 3 (methionine), -CH 2 SCH 3 (S- methylcysteine), (1 /-/-imidazol-4-yl)methyl- (histidine), (1 /-/-indol-3-yl)methyl- (thryptophan), -CH 2 NH 2 (2,3-diaminopropanoic acid), and -(CH 2 ) 2 NH 2 (2,4- diaminobutanoic acid),

R 8 represents hydrogen or a group selected from:

Ci-C 3 -alkyl, Ci-C 3 -haloalkyl, C 2 -C 3 -hydroxyalkyl, C 3 -C 4 -cycloalkyl,

(C 3 -C 4 -cycloalkyl)-(Ci-C 3 -alkyl)-, and (Ci-C 3 -alkoxy)-(C 2 -C 3 -alkyl)-, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a pharmaceutically acceptable salt, or a mixture of same.

In accordance to an embodiment of the first aspect, component A is a Bub1 kinase inhibitor of formula (I) supra, wherein,

V, W, Y and Z independently of each other represent CH or CR 2 , wherein one of V, W,

Y and Z represents CR 2

or,

V represents N, and W, Y and Z independently of each other represent CH or CR 2 ,

R 1 represents a group selected from :

Ci-C 3 -alkyl, Ci-C 3 -haloalkyl, and C 3 -C4-cycloalkyl,

R 2 represents, independently of each other, halogen or a group selected from: Ci-C 3 -alkyl, C 3 -C 4 -cycloalkyl, Ci-C 3 -haloalkyl, Ci-C 3 -alkoxy,

Ci-C 3 -haloalkoxy, -N(H)C(=0)H,

-N(H)C(=0)-(Ci-C 3 -hydroxyalkyl),

-N(H)C(=0)-(Ci-C 3 -alkyl)-(Ci-C 3 -alkoxy), -N(H)C(=0)-phenyl, -N(H)C(=0)-(C 3 -C 4 -cycloalkyl),

-N(H)C(=0)-(Ci-C 3 -alkyl)-(C 3 -C 4 -cycloalkyl), and -N(H)C(=0)N(H)R 8 , said -N(H)C(=0)-phenyl being optionally substituted at the phenyl ring, one, two or three times, identically or differently, with a substituent selected from:

halogen, hydroxy, cyano, Ci-C 4 -alkyl, Ci-C 4 -haloalkyl,

Ci-C 4 -alkoxy, Ci-C 4 -haloalkoxy, C 3 -C -cycloalkyl, and C 3 -C -cycloalkyloxy,

said -N(H)C(=0)-(C 3 -C -cycloalkyl) being optionally substituted at the C 3 -C -cycloalkyl ring with a substituent selected from:

fluorine, chlorine, trifluoromethyl, and methoxy, R 3 represents a group selected from :

C2-C6-hydroxyalkyl, and R 4 ,

said C 2 -C 6 -hydroxyalkyl group being optionally substituted with one, two or three halogen atoms selected from:

fluorine, and chlorine,

R 4 represents -(C2-C 6 -alkyl)-OC(=0)-C(H)(R 5 )-N(H)C(=0)-C(H)(R 7 )-NH 2 , in which C2-C6-alkyl is optionally substituted with one, two or three halogen atoms selected from:

fluorine, and chlorine,

R 5 and R 7 independently of each other represent a group selected from:

-CH 3 (alanine), -C(H)(CH 3 ) 2 (valine), -(CH 2 ) 2 CH 3 (norvaline), -(CH 2 ) 3 NH 2 (ornithine), -

(CH 2 ) 4 NH 2 (lysine), and -(CH 2 ) 3 N(H)C(=NH)NH 2 (arginine), R 8 represents hydrogen or a group selected from:

Ci-C 3 -alkyl, Ci-C 3 -haloalkyl, C 2 -C 3 -hydroxyalkyl, C 3 -C -cycloalkyl,

(C 3 -C 4 -cycloalkyl)-(Ci-C 3 -alkyl)-, and (Ci-C 3 -alkoxy)-(C 2 -C 3 -alkyl)-, 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 accordance to an embodiment of the first aspect, component A is a Bub1 kinase inhibitor of formula (I) supra, wherein,

V, W, Y and Z independently of each other represent CH or CR 2 , wherein one of V, W, Y and Z represents CR 2 or,

V represents N, and W, Y and Z independently of each other represent CH or CR 2 , R 1 represents a Ci-C 3 -alkyl group,

R 2 represents, independently of each other, halogen or a group selected from: Ci-C 3 -alkyl, C3-C 4 -cycloalkyl, Ci-C 3 -haloalkyl, Ci-C 3 -alkoxy,

R 3 represents a group selected from :

C2-C6-hydroxyalkyl, and R 4 ,

R 4 represents -(C2-C 6 -alkyl)-OC(=0)-C(H)(R 5 )-N(H)C(=0)-C(H)(R 7 )-NH 2 , R 5 and R 7 independently of each other represent a group selected from :

-CH 3 (alanine), and -(CH 2 ) 4 NH 2 (lysine), 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 accordance to an embodiment of the first aspect, component A is a Bub1 kinase inhibitor of formula (I) supra, wherein,

V, W, Y and Z independently of each other represent CH or CR 2 , wherein one of V, W, Y and Z represents CR 2

or,

V represents N, and W, Y and Z independently of each other represent CH or CR 2 , R 1 represents a methyl group,

R 2 represents, independently of each other, fluorine, chlorine or a group selected from:

methyl, cyclopropyl, difluoromethyl, methoxy, and -N(H)C(=0)-CH 3 ,

R 3 represents a group selected from :

-(CH 2 ) 2 OH, and R 4 , R 4 represents -(CH 2 )2-OC(=0)-C(H)(R 5 )-N(H)C(=0)-C(H)(R 7 )-NH 2 ,

R 5 represents -CH 3 (alanine),

R 7 represents -(CH 2 ) 4 NH2 (lysine), 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 accordance to an embodiment of the first aspect, component A is a Bub1 kinase inhibitor of formula (I) supra, wherein,

V, W, Y and Z independently of each other represent CH or CR 2 , wherein one of V, W, Y and Z represents CR 2

or,

V represents N, and W, Y and Z independently of each other represent CH or CR 2 , R 1 represents a methyl group,

R 2 represents, independently of each other, fluorine, chlorine or a group selected from:

methyl, cyclopropyl, difluoromethyl, methoxy, -N(H)C(=0)-CH 3 , -N(H)C(=0)-cyclopropyl, and -N(H)C(=0)N(H)-cyclopropyl,

R 3 represents a -(CH 2 ) 2 OH 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.

In accordance to an embodiment of the first aspect, component A is a Bub1 kinase inhibitor of formula (I) supra, selected from the group consisting of :

2-{4-[(3-{4-[(3-chloropyridin-4-yl)amino]-5-methoxypyrimidin -2-yl}-1 /-/-indazol-1 - yl)methyl]-3,5-difluorophenoxy}ethanol ,

2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(2-methylpyridin-4-yl)am ino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethanol ,

2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl)a mino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethanol ,

N-{4-[(2-{1 -[2,6-difluoro-4-(2-hydroxyethoxy)benzyl]-1 /-/-indazol-3-yl}-5- methoxypyrimidin-4-yl)amino]pyridin-2-yl}acetamide ,

2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methylpyridin-4-yl)am ino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethanol ,

2-{3,5-difluoro-4-[(3-{4-[(3-fluoropyridin-4-yl)amino]-5-met hoxypyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethanol ,

2-(4-{[3-(4-{[2-(difluoromethyl)pyridin-4-yl]amino}-5-methox ypyrimidin-2-yl)-1 /-/-indazol- 1 -yl]methyl}-3,5-dif luorophenoxy)ethanol ,

2-{4-[(3-{4-[(2,5-dimethylpyridin-4-yl)amino]-5-methoxypyrim idin-2-yl}-1 /-/-indazol-1 - yl)methyl]-3,5-difluorophenoxy}ethanol ,

2-{4-[(3-{4-[(3-cyclopropylpyridin-4-yl)amino]-5-methoxypyri midin-2-yl}-1 /-/-indazol-1 - yl)methyl]-3,5-difluorophenoxy}ethanol ,

2-{1 -[2,6-difluoro-4-(2-hydroxyethoxy)benzyl]-1 /-/-indazol-3-yl}-4-[(2-methylpyrimidin-4- yl)amino]pyrimidin-5-ol

2-{3,5-difluoro-4-[(3-{4-[(2-fluoropyridin-4-yl)amino]-5-met hoxypyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethanol ,

2-{3,5-difluoro-4-[(3-{4-[(5-fluoro-2-methylpyridin-4-yl)ami no]-5-methoxypyrimidin-2-yl}- 1 H-indazol-1 -yl)methyl]phenoxy}ethanol ,

2-[3,5-difluoro-4-({3-[5-methoxy-4-(pyrimidin-4-ylamino)pyri midin-2-yl]-1 H-indazol-1 - yl}methyl)phenoxy]ethanol

/V-{4-[(2-{1 -[2,6-difluoro-4-(2-hydroxyethoxy)benzyl]-1 H-indazol-3-yl}-5- methoxypyrimidin-4-yl)amino]-5-methylpyridin-2-yl}acetamide ,

2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(2-methylpyridin-4-yl )amino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl L-lysyl-L-alaninate, salt with trifluoroacetic acid , 2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(2-methylpyridin-4-yl)am ino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl L-lysyl-L-alaninate ,

2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl)a mino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl L-lysyl-L-alaninate, salt with trifluoroacetic acid , 2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl)a mino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl L-lysyl-L-alaninate,

N-{4-[(2-{1 -[2,6-difluoro-4-(2-hydroxyethoxy)benzyl]-1 H-indazol-3-yl}-5-methoxy- pyrimidin-4-yl)amino]pyridin-2-yl}cyclopropanecarboxamide ,

1 -cyclopropyl-3-{4-[(2-{1 -[2,6-difluoro-4-(2-hydroxyethoxy)benzyl]-1 H-indazol-3-yl}-5- methoxypyrimidin-4-yl)amino]pyridin-2-yl}urea ,

2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxy-2-methylpyrid in-4-yl)amino]pyrimi^ yl}-1 H-indazol-1 -yl)methyl]phenoxy}ethanol ,

2- {3,5-difluoro-4-[(3-{5-methoxy-4-[(5-methoxy-2-m

yl}-1 H-indazol-1 -yl)methyl]phenoxy}ethanol ,

3- {4-[(3-{4-[(2,5-dimethylpyridin-4-yl)amino]-5-methoxypyrimid in-2-yl}-1 H-indazol-1 - yl)methyl]-3,5-difluorophenoxy}propan-1 -ol ,

3-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl)a mino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}propan-1 -ol ,

3-{3,5-difluoro-4-[(3-{5-methoxy-4-[(2-methylpyrimidin-4-yl) amino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}propan-1 -ol ,

(2R)-3-{3,5-difluoro-4-[(3-{5-methoxy-4-[(2-methylpyrimidin- 4-yl)amino]pyrimidin-2-yl}- 1 H-indazol-1 -yl)methyl]phenoxy}-2-methylpropan-1 -ol ,

(2R)-3-{4-[(3-{4-[(2,5-dimethylpyridin-4-yl)amino]-5-methoxy pyrimidin-2-yl}-1 H-indazol- 1 -yl)methyl]-3,5-difluorophenoxy}-2-methylpropan-1 -ol ,

N-[4-({2-[1 -(2,6-difluoro-4-{[(2R)-3-hydroxy-2-methylpropyl]oxy}benzyl) -1 H-indazol-3- yl]-5-methoxypyrimidin-4-yl}amino)pyridin-2-yl]acetamide ,

(2R)-3-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4 -yl)amino]pyrimidin-2-yl}- 1 H-indazol-1 -yl)methyl]phenoxy}-2-methylpropan-1 -ol ,

3-{3,5-difluoro-4-[(3-{5-methoxy-4-[(2-methylpyridin-4-yl)am ino]pyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}propan-1 -ol ,

2-{4-[(3-{4-[(2,6-dimethylpyrimidin-4-yl)amino]-5-methoxypyr imidin-2-yl}-1 H-indazol-1 - yl)methyl]-3,5-difluorophenoxy}ethanol ,

2-{3,5-difluoro-4-[(3-{4-[(2-fluoropyridin-4-yl)amino]-5-met hoxypyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl D-lysyl-L-alaninate, salt with trifluoroacetic acid , 2-{3,5-difluoro-4-[(3-{4-[(2-fluoropyridin-4-yl)amino]-5-met hoxypyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl D-lysyl-L-alaninate ,

2-{3,5-difluoro-4-[(3-{4-[(2-fluoropyridin-4-yl)amino]-5-met hoxypyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl L-lysyl-L-alaninate, salt with trifluoroacetic acid , and 2-{3,5-difluoro-4-[(3-{4-[(2-fluoropyridin-4-yl)amino]-5-met hoxypyrimidin-2-yl}-1 H- indazol-1 -yl)methyl]phenoxy}ethyl L-lysyl-L-alaninate, or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a pharmaceutically acceptable salt, or a mixture of same. In accordance to a preferred embodiment of the first aspect, component A is a Bub1 kinase inhibitor of formula (I) supra, wherein the component A is 2-{3,5-difluoro-4-[(3-{5- methoxy-4-[(3-methoxypyridin-4-yl)amino]pyrimidin-2-yl}-1 /-/-indazol-1 - yl)methyl]phenoxy}ethanol,

5 or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a pharmaceutically acceptable salt, or a mixture of same.

In accordance to a more preferred embodiment of the first aspect, component A is 2- {3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl)ami no]pyrimidin-2-yl}-1 AVICI indazol-1 -yl)methyl]phenoxy}ethanol,

or a pharmaceutically acceptable salt thereof.

In accordance to a more preferred embodiment of the first aspect, component A is 2- {3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4-yl)ami no]pyrimidin-2-yl}-1 H- 15 indazol-1 -yl)methyl]phenoxy}ethanol.

In accordance to a preferred embodiment of the first aspect, component B is docetaxel. In accordance to a preferred embodiment of the first aspect, component B is paclitaxel.

20

In accordance to an embodiment, the cancer is breast cancer and/or metastases thereof, such as Triple-negative breast cancer (TNBC), antiestrogen resistant breast cancer and aromatase inhibitor-resistant breast cancer and/or metastases thereof.

25 In accordance to an embodiment, the cancer is prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC) and/or metastases thereof.

In accordance to a preferred embodiment, the cancer is Triple-negative breast cancer (TNBC) and/or metastases thereof.

30

In accordance to a preferred embodiment, the cancer is Castration-Resistant Prostate Cancer (CRPC) and/or metastases thereof.

In accordance to a preferred embodiment, the cancer is Non-Small Cell Lung Cancer 35 (NSCLC) and/or metastases thereof. In accordance to a preferred embodiment, the cancer is melanoma and/or metastases thereof.

In accordance to a preferred embodiment, the cancer is colorectal cancer, such as colorectal adenocarcinoma and/or metastases thereof.

In accordance to a preferred embodiment, the cancer is glioblastoma/neuroglioma and/or metastases thereof. In accordance to a preferred embodiment, the cancer is ovarian cancer, such as ovarian adenocarcinoma and/or metastases thereof.

In accordance to a preferred embodiment, the cancer is cervical cancer, such as cervical adenocarcinoma and/or metastases thereof.

In accordance to a preferred embodiment, the cancer is gastric (stomach) cancer and/or metastases thereof.

In accordance to a preferred embodiment, the cancer is pancreatic cancer, such as pancreatic carcinoma and/or metastases thereof.

In accordance to a preferred embodiment, the cancer is bladder cancer, such as bladder transitional cell carcinoma and/or metastases thereof. DETAILED DESCRIPTION A. Definitions

The combinations comprising at least two components, component A and component B, as decribed and defined herein, are also referred to as "combinations of the present invention".

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 R 1 , R 2 , R 3 , R 4 , R 5 , R 7 , R 8 V, W, Y and/or Z occur more than one time for any compound of formula (I) each definition of R\ R 2 , R 3 , R 4 , R 5 , R 7 , R 8 , V, W, Y and Z is independent.

Should a constituent be composed of more than one part, e.g. Ci-C 4 -alkoxy-Ci-C 4 -alkyl- , the position of a possible substituent can be at any of these parts at any suitable position. A hyphen at the beginning or at the end of the constituent marks the point of attachment to the rest of the molecule. Should a ring be substituted the substitutent could be at any suitable position of the ring, also on a ring nitrogen atom if suitable. The term "comprising" when used in the specification includes "consisting of".

If it is referred to "above or "supra", alone or in expressions such as "as mentioned above", "mentioned above", or "as defined supra", within the description it is referred to any of the disclosures made within the specification in any of the preceding pages.

If it is referred to "herein", alone or in expressions such as "as mentioned herein", "mentioned herein", or "as described herein" within the description it is referred to any of the disclosures made within the specification in any of the preceding or subsequent 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-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. a methyl, ethyl, propyl, butyl, pentyl, hexyl, 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 or 4 carbon atoms ("Ci-C 4 -alkyl"), e.g. a methyl, ethyl, propyl, butyl, iso-propyl, iso-butyl, sec-butyl, tert-butyl 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-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. Said Ci-C 6 -haloalkyl group is, for example, -CF 3 , - CHF 2 , -CH 2 F, -CF2CF3, -CH2CH2F, -CH2CHF2, -CH2CF3, -CH2CH2CF3, or -CH(CH 2 F) 2 .

The term "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. Particularly preferred is "Ci -C 4 -alkoxy" e.g. a methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, iso-butoxy, tert-butoxy or sec-butoxyor an isomer thereof. More preferred is "Ci-alkoxy", i.e. methoxy.

The term "Ci-C 4 -haloalkoxy" is to be understood as meaning a linear or branched, saturated, monovalent Ci -C 4 -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-C 4 -haloalkoxy group is, for example, - OCF3, -OCHF2, -OCH2F, -OCF2CF3, or -OCH2CF3.

The term "Ci-C 6 -hydroxyalkyl" 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. Preferred is "Ci-C3-hydroxyalkyl", e.g. a hydroxymethyl, 1 -hydroxyethyl, 2-hydroxyethyl, 1 ,2-dihydroxyethyl, 3-hydroxypropyl, 2-hydroxypropyl, 2,3-dihydroxypropyl, 1 ,3- dihydroxypropan-2-yl. The term "C2-C 6 -hydroxyalkyl" is to be understood as meaning a linear or branched, saturated, monovalent hydrocarbon group having 2, 3, 4, 5, or 6 carbon atoms, in which one or more hydrogen atoms is replaced by a hydroxy group, e.g. a 2-hydroxyethyl, 3- hydroxypropyl, 2-hydroxypropyl, 2,3-dihydroxypropyl, 3-hydroxy-2-methyl-propyl, 2- hydroxy-2-methyl-propyl. Preferred is "C2-C 4 -hydroxyalkyl", more preferred is "C 2 - hydroxyalkyl", i.e. a 2-hydroxyethyl group.

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 ("C 3 -C 6 - cycloalkyi"). Said C3-C 6 -cycloalkyl group is for example, a monocyclic hydrocarbon ring, e.g. a cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl ring.

The term "C3-C 6 -cycloalkyloxy" is to be understood as meaning a saturated, monovalent, monocyclic hydrocarbon group of formula -O-cycloalkyI, in which the term "cycloalkyi" is defined supra, e.g. a. a cyclopropyloxy, cyclobutyloxy, cyclopentyloxy or cyclohexyloxy group.

The term "Ci-C 6 ", as used throughout this text, e.g. in the context of the definition of "Ci- Ce-alkyl", "Ci-C 6 -haloalkyl", "Ci-C 6 -hydroxyalkyl", "Ci-C 6 -alkoxy", is to be understood as meaning an alkyl group having a finite number of carbon atoms of 1 to 6, i.e. 1 , 2, 3, 4, 5, or 6 carbon atoms. It is to be understood further that said term "Ci-Ce" is to be interpreted as any sub-range comprised therein, e.g. Ci-C 6 , C 2 -C 5 , C 3 -C 4 , C1-C2, C1-C3 , C1-C4 , C1-C5 ; particularly C1-C2 , C1-C3 , C1-C4 , C1-C5, Ci-C 6; more particularly C1-C4 ; in the case of "Ci-C 6 -haloalkyl" or "Ci-C 6 -haloalkoxy" even more particularly C1-C2.

The term "C 2 -C 6 ", as used throughout this text, e.g. in the context of the definition of "C 2 - Ce-hydroxyalkyl", is to be understood as meaning a hydroxyalkyl group having a finite number of carbon atoms of 2 to 6, i.e. 2, 3, 4, 5, or 6 carbon atoms. It is to be understood further that said term "C 2 -C 6 " is to be interpreted as any sub-range comprised therein, e.g. C 2 -C 6 , C 2 -C 5 , C3-C4, particularly C 2 -C 3 , C 2 -C 4 , C 2 -C 5 , C 2 -C 6 .

Further, as used herein, the term "C3-C 6 ", as used throughout this text, e.g. in the context of the definition of "C3-C 6 -cycloalkyl", is to be understood as meaning a cycloalkyi group having a finite number of carbon atoms of 3 to 6, i.e. 3, 4, 5 or 6 carbon atoms. It is to be understood further that said term "C3-C 6 " is to be interpreted as any sub-range comprised therein, e.g. C 3 -C 6 , C 4 -C 5 , C3-C5 , C3-C4 , C 4 -C 6 , C 5 -C 6 ; particularly

The term "substituted" means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. The term "optionally substituted" means optional substitution with the specified groups, radicals or moieties.

Ring system substituent means a substituent attached to an aromatic or nonaromatic ring system which, for example, replaces an available hydrogen on the ring system.

As used herein, the term "one or more", e.g. in the definition of the substituents of the compounds of the general formulae of the present invention, is understood as meaning "one, two, three, four or five, particularly one, two, three or four, more particularly one, two or three, even more particularly one or two".

The term "V, W, Y and Z independently of each other represent CH or CR 2 , wherein one of V, W, Y and Z represents CR 2 ", is to be understood as meaning that at least one of V, W, Y and Z represents CR 2 , and the remaining, independently from each other, represent CH or CR 2 , as it is known to a skilled person. For example, according to certain embodiments of the invention, V, W, Y and Z independently of each other represent CH or CR 2 , wherein one of V, W, Y and Z represents CR 2 and the remaining represent CH; according to other embodiments of the invention, V, W, Y and Z independently of each other represent CH or CR 2 , wherein two of V, W, Y and Z, independently of each other, represent CR 2 and the remaining represent CH; still according to other embodiments of the invention, V, W, Y and Z independently of each other represent CH or CR 2 , wherein three of V, W, Y and Z, independently of each other, represent CR 2 and the remaining represents CH, for example.

The invention also includes all suitable isotopic variations of a compound (i.e. component A, B or C(when present)) used in the combination of the present invention. An isotopic variation of a compound is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually or predominantly found in nature. Examples of isotopes that can be incorporated into a compound used in the combination of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, 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, 129 l and 131 1, respectively. Certain isotopic variations of a compound used in the combination of the present invention, for example, those in which one or more radioactive isotopes such as 3 H or 14 C are incorporated, are useful in drug and/or substrate tissue distribution studies. Tritiated and carbon-14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence is preferred in some circumstances. Isotopic variations of a compound of the invention can generally be prepared by conventional procedures known by a person skilled in the art such as by the illustrative methods or by the preparations described in the examples hereafter using appropriate isotopic variations of suitable reagents.

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 used in the combination of this invention optionally contain one or more asymmetric centre, depending upon the location and nature of the various substituents desired. Asymmetric carbon atoms is present in the (R) or (S) configuration, resulting in racemic mixtures in the case of a single asymmetric centre, and diastereomeric mixtures in the case of multiple asymmetric centres. In certain instances, asymmetry may also be present due to restricted rotation about a given bond, for example, the central bond adjoining two substituted aromatic rings of the specified compounds. The compounds used in the combination of the present invention optionally contain sulphur atoms which are asymmetric, such as an asymmetric sulfoxide, of structure: , for example, in which * indicates atoms to which the rest of the molecule can be bound.

Substituents on a ring may also be present in either cis or trans form. It is intended that all such configurations (including enantiomers and diastereomers), are included within the scope of the present invention.

Preferred compounds used in the combination 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 used in the combination 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 used in the combination 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 used in the combination 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 used in the combination of the present invention may exist as tautomers.

The present invention includes all possible tautomers of the compounds used in the combination of the present invention as single tautomers, or as any mixture of said tautomers, in any ratio.

Further, the compounds used in the combination 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 used in the combination as disclosed herein, such as metabolites, hydrates, solvates, prodrugs, salts, in particular pharmaceutically acceptable salts, and co-precipitates.

The compounds used in the combination of the present invention can exist as a hydrate, or as a solvate, wherein the compounds 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 used in the combination 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 used in the combination of the present invention may be, for example, an acid-addition salt of a compound 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 used in the combination 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, aralkyi halides like benzyl and phenethyl bromides and others. Those skilled in the art will further recognise that acid addition salts of the 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 used in the combination of the invention are prepared by reacting the compounds with the appropriate base via a variety of known methods.

The present invention includes all possible salts of the compounds used in the combination 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 CF 3 COOH", "x Na + ", for example, are to be understood as not a stoichiometric specification, but solely as a salt form. This applies analogously to cases in which synthesis intermediates or example compounds or salts thereof have been obtained, by the preparation and/or purification processes described, as solvates, such as hydrates with (if defined) unknown stoichiometric composition. As used herein, the term "in vivo hydrolysable ester" is understood as meaning an in vivo hydrolysable ester of a compound used in the combination 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-Ce alkanoyloxymethyl esters, e.g. pivaloyloxymethyl, phthalidyl esters, C 3 -C 8 cycloalkoxy-carbonyloxy-Ci-C 6 alkyl esters, e.g. 1 -cyclohexylcarbonyloxyethyl ; 1 ,3-dioxolen-2-onylmethyl esters, e.g. 5-methyl-1 ,3- dioxolen-2-onylmethyl ; and Ci-C 6 -alkoxycarbonyloxyethyl esters, e.g. 1 - methoxycarbonyloxyethyl, and may be formed at any carboxy group in the compounds used in the combination of this invention. An in vivo hydrolysable ester of a compound used in the combination 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 used in the combination 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 used in the combination 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 component A and component B 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 component A and the said component B 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 component A and the said component B 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 component A and the said component B (and optionally component C) are present in more than one unit. One example of a non-fixed combination or kit-of-parts is a combination wherein the said component A and the said component B (and optionally component C) are present separately, for example in different and separate pharmaceutical compositions. The components of the non-fixed combination or kit-of-parts may be administered separately, sequentially, simultaneously, concurrently or chronologically staggered.

Any such combination is covered by the present invention.

As defined herein the term "standard of care drug(s)" is meant to be understood a drug selected froma taxane, such as docetaxel or paclitaxel, or combinations thereof.

As used herein the term "resistant" is applied interchangeably with "refractory".

The term " resistant" or "insensitive" to "treatment with stardard of care drugs" is meant to define a cancer disease, particularly breat cancer such as, Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC) or Non-Small Cell Lung Cancer (NSCLC) and/or metastases thereof, in which the treatment with a drug selected from a taxane, such as docetaxel or paclitaxel, or combinations thereof,

is not therapeutically effective, for failing to:

- reduce the growth of a tumor and/or metastasis thereof or even eliminate the tumor and/ or metastasis thereof,

- prevent regrowth of a tumor after initial response,

- provide for a longer survival time, and/or

- provide a longer time for tumor progression.

The expression "antiestrogen-resistant breast cancer" refers to breast cancers for which treatment with antiestrogens, such as tamoxifen, raloxifene, toremifene, and fulvestrant, is not therapeutically effective, for failing to:

- reduce the growth of a tumor and/or metastasis thereof or even eliminate the tumor and/ or metastasis thereof, - prevent regrowth of a tumor after initial response,

- provide for a longer survival time, and/or

- provide a longer time for tumor progression.

The expression "aromatase inhibitor-resistant breast cancer", refers to breast cancers for which treatment with aromatase inhibitors, such as Anastrozole (Arimidex®), Exemestane (Aromasin®), and Letrozole (Femara®), is not therapeutically effective, for failing to:

- reduce the growth of a tumor and/or metastasis thereof or even eliminate the tumor and/ or metastasis thereof,

- prevent regrowth of a tumor after initial response,

- provide for a longer survival time, and/or

- provide a longer time for tumor progression.

The resistance and/or insensitivity may be intrinsic (to the patient) or acquired.

The methods, combinations, uses and kits of the present invention cover both intrinsic and acquired resistance and/or insensitivity to standard of care drugs as defined herein.

It is further to be understood that embodiments disclosed herein are not meant to be understood as individual embodiments which would not relate to one another. Features discussed with one embodiment or aspect of the invention are meant to be disclosed also in connection with other embodiments or aspects of the invention shown herein. If, in one case, a specific feature is not disclosed with one embodiment or aspect of the invention, but with another, the skilled person would understand that does not necessarily mean that said feature is not meant to be disclosed with said other embodiment or aspect of the invention. The skilled person would understand that it is the gist of this application to disclose said feature also for the other embodiment or spect of the invention, but that just for purposes of clarity and to keep the length of this specification manageable. It is further to be understood that the content of the prior art documents referred to herein is incorporated by reference, e.g., for enablement purposes, namely when e.g. a method is discussed details of which are described in said prior art document. This approach serves to keep the length of this specification manageable. Component A of the Combination

Component A can be selected from inhibitors of BUB1 -kinase or a pharmaceutically acceptable salt, solvate, hydrate or stereoisomer thereof specifically or generically disclosed e.g. in the publications as mentioned above which are incorporated herein by reference.

In an embodiment Component A is selected from the group of Bub1 inhibitors generically or specifically disclosed in WO 2016/042084, which are incorporated by reference herein.

In accordance with a preferred embodiment, component A is is 2-{3,5-difluoro-4-[(3-{5- methoxy-4-[(3-methoxypyridin-4-yl)amino]pyrimidin-2-yl}-1 /-/-indazol-1 - yl)methyl]phenoxy}ethanol,

or a stereoisomer, a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, in particular a pharmaceutically acceptable salt, or a mixture of same.

In a more preferred embodiment, said component A is 2-{3,5-difluoro-4-[(3-{5-methoxy- 4-[(3-methoxypyridin-4-yl)amino]pyrimidin-2-yl}-1 /-/-indazol-1 -yl)methyl]phenoxy}ethanol or a pharmaceutically acceptable salt thereof.

In a more preferred embodiment, component A is 2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3- methoxypyridin-4-yl)amino]pyrimidin-2-yl}-1 /-/-indazol-1 -yl)methyl]phenoxy}ethanol.

In other embodiment, component A is selected from the group of Bub1 inhibitors described generically or specifically in:

WO2013/050438

WO 2013/092512

WO 2013/167698

WO 2014/147144

WO 2014/147203

WO 2014/147204

WO2014/202586

WO 2014/202590

WO 2014/202588 WO 2014/ 202583

WO 2015/193339

WO 2016/041925

WO 2016/042080

WO 2106/ 042081

WO 2016/120196,

WO2016/202755

PCT/EP2015/063527 (WO2015/193339)

PCT/EP2016/068279 (WO2017/021348), or

EP15200590.6 or

WO2017/102649,

which are incorporated herein by reference in their entirety.

Compounds of formula (I) as described and defined herein can be prepared according to the preparation methods contained in WO 2016/042084 which is incorporated herein by reference in its entirety. The BUB1 -inhibitors mentioned in the prior art as well as in the lists above have been disclosed for the treatment or prophylaxis of different diseases, especially cancer.

The specific compounds of the lists as disclosed above are preferred as being component A of the combination, most preferred are the compounds used in the experimental section.

The synergistic behavior of a combination of the present invention is demonstrated herein with a Bub1 inhibitor specifically disclosed as example 2-1 -3 in WO 2016/042084, referrred to as Compound A1 (or as Compd A1 ) below.

A preferred embodiment of the present invention covers combinations comprising the Compound A1 or a pharmaceutically acceptable salt thereof and a taxane selected from docetaxel and paclitaxel. It is to be understood that the present invention relates also to any combination of the embodiments of component A described above.

Component A may be administered by the oral, intravenous, topical, local installations, intraperitoneal or nasal route.

Preferably Component A is administered intravenously, intraperitoneally or orally.

Compound A1 is administered preferably orally.

Component B of the Combination

Component B is an anti-hyperproliferative, cytotoxic and/or cytostatic agent selected from a taxane, such as docetaxel or paclitaxel, or combinations thereof. Suitable dose(s), administration regime(s) and administration route(s) for taxanes include those described in the NCCN Clinical Practice Guidelines in Oncology (NCCN guidelines), in particular in the NCCN Guidelines for Breast Cancer version 2.2016, the NCCN Guidelines for Prostate Cancer version 2.2017 and the NCCN Guidelines for Non-Small Cell Lung Cancer version 4.2017, which are included herein by reference in their entirety.

The term "cytotoxic" refers to an agent which can be administered to kill or eliminate a cancer cell. The term "cytostatic" refers to an agent which can be administered to restrain tumor proliferation rather than induce cytotoxic cytoreduction yielding an elimination of the cancer cell from the total viable cell population of the patient. The term "anti-hyperproliferative" refers to an agent which can inhibit the survival or multiplication of the tumor cells with high proliferation rate.

The chemotherapeutic agents described herein, e.g., docetaxel and paclitaxel are considered cytotoxic, cytostatic agent, or anti-hyper-proliferative agents depending on individual tumor types. These anti-hyperproliferative, cytotoxic and cytostatic agents have gained wide spread use as chemotherapeutics in the treatment of various cancer types and are well known. Component B may be administered by the oral, intravenous, topical, local installations, intraperitoneal or nasal route. Docetaxel is sold under the tradename Taxotere® by Sanofi-Aventis (1 ,7β,10β- trihydroxy-9-oxo-53,20-epoxytax-1 1 -ene-2a,4,13a-triyl 4-acetate 2-benzoate 13- {(2 ,3S)-3-[(fert-butoxycarbonyl)amino]-2-hydroxy-3-phenylpropan oate}, CAS No. 1 14977-28-5). Docetaxel belongs to the taxanes chemotherapy drug class and is a semi-synthetic analogue of paclitaxel (Taxol®). It is an anti-mitotic chemotherapy medication that works by interfering with cell division. While not bound by a theory, the cytotoxic activity of docetaxel is believed to be exerted by promoting and stabilising microtubule assembly, while preventing physiological microtubule depolymerisation/disassembly in the absence of GTP. This leads to a significant decrease in free tubulin, needed for microtubule formation and results in inhibition of mitotic cell division between metaphase and anaphase, preventing further cancer cell progeny. Because microtubules do not disassemble in the presence of docetaxel, they accumulate inside the cell and cause initiation of apoptosis. Docetaxel is administered by intravenous injection or by other appropriate infusion techniques.

Paclitaxel is sold under the tradename Taxol® by the Bristol-Myers Squibb Company. Paclitaxel ((2a,4a,53,73,103,13a)-4,10-bis(acetyloxy)-13-{[(2R,3S)- 3-(benzoylamino)- 2-hydroxy-3-phenylpropanoyl]oxy}- 1 ,7-dihydroxy-9-oxo-5,20-epoxytax-1 1 -en-2-yl benzoate - CAS No: 33069-62-4) has the empirical formula C47H51 N014 and a molecular weight of 853.9. It is highly lipophilic in water. Paclitaxel is an antimicrotubule agent that promotes the assembly of microtubles from tubulin dimers and stabilizes microtubules by preventing depolymerization. While not bound by a theory, it is believed that this stability results in the inhibition in the normal dynamic reorganization of the microtubule network that is essential for vital interphase and mitotic cellular functions. Also, paclitaxel is believed to induce abnormal arrays or bundles of microtubules throughout the cell cycle and multiple asters of microtubules during mitosis. Paclitaxel is administered by intravenous injection or by other appropriate infusion techniques.

Protein-bound paclitaxel (Tradename: Abraxane® ) is an injectable formulation of paclitaxel, a mitotic inhibitor drug used in the treatment of breast cancer, lung cancer (NSCLC) and pancreatic cancer. In this formulation, paclitaxel is bonded to albumin as a delivery vehicle. It is also called nab-paclitaxel (with the "nab" syllable derived from "nanoparticle albumin-bound") or paclitaxel albumin-bound. Unless specified otherwise, for the purpose of the present invention the term "paclitaxel" is meant to cover both paclitaxel and protein-bound paclitaxel.

Accordingly, in certain embodiments of the present invention component B is paclitaxel or protein-bound paclitaxel and is collectively referred to as "paclitaxel".

Alternatively, in certain embodiments of the present invention component B is paclitaxel only (i.e. ((2a,4a,53,73,103,13a)-4,10-bis(acetyloxy)-13-{[(2R,3S)- 3-(benzoylamino)-2- hydroxy-3-phenylpropanoyl]oxy}- 1 ,7-dihydroxy-9-oxo-5,20-epoxytax-1 1 -en-2-yl benzoate - CAS No : 33069-62-4) .

Alternatively!, in certain embodiments of the present invention component B is protein- bound paclitaxel only.

The term "component C" being at least one pharmaceutical agent includes the effective compound itself as well as its pharmaceutically acceptable salts, solvates, hydrates or stereoisomers as well as any composition or pharmaceutical formulation comprising such effective compound or its pharmaceutically acceptable salts, solvates, hydrates or stereoisomers. A list of such readily available agents is being provided further below.

The components may be administered independently of one another by the oral, intravenous, topical, local installations, intraperitoneal or nasal route.

Component B preferably is administered by the more appropriate route within the knowledge of the skilled person. Suitable route(s) are included in NCCN Guidelines for NSCLC Version 2.2013, which is included herein by reference in its entirety. Component C being administered as the case may be.

Components of this invention can be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatin, 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, coloring agents, and flavoring agents such as peppermint, oil of wintergreen, or cherry flavoring, 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, flavoring and coloring agents described above, may also be present.

Components of this invention can 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 flavoring agents. Oily suspensions can 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 coloring agents; one or more flavoring agents; and one or more sweetening agents such as sucrose or saccharin. Syrups and elixirs can 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 flavoring and coloring agents.

Components of this invention can also be administered parenterally, that is, subcutaneously, intravenously, intraocularly, intrasynovially, intramuscularly, or interperitoneally, as injectable dosages of the component 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 minimize 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 can 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 can 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. Components of the invention can also be administered in the form of suppositories for rectal administration of the drug. These components can be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are, for example, cocoa butter and polyethylene glycol.

Another formulation employed in the methods of the present invention employs transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art (see, e.g., US Patent No. 5,023,252, issued June 1 1 , 1991 , incorporated herein by reference). Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents. Controlled release formulations for parenteral administration include liposomal, polymeric microsphere and polymeric gel formulations that are known in the art.

It can be desirable or necessary to introduce a component of the present invention to the patient via a mechanical delivery device. The construction and use of mechanical delivery devices for the delivery of pharmaceutical agents is well known in the art. Direct techniques for, for example, administering a drug directly to the brain usually involve placement of a drug delivery catheter into the patient's ventricular system to bypass the blood-brain barrier. One such implantable delivery system, used for the transport of agents to specific anatomical regions of the body, is described in US Patent No. 5,01 1 ,472, issued April 30, 1991 .

The compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired. Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized. Such ingredients and procedures include those described in the following references, each of which is incorporated herein by reference: Powell, M.F. et al, "Compendium of Excipients for Parenteral Formulations " PDA Journal of Pharmaceutical Science & 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 charcoal); aerosol propellents (examples include but are not limited to carbon dioxide, CCI2F2, 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) colorants (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) flavorants (examples include but are not limited to anise oil, cinnamon oil, cocoa, menthol, orange oil, peppermint oil and vanillin); humectants (examples include but are not limited to glycerol, propylene glycol and sorbitol); levigating agents (examples include but are not limited to mineral oil and glycerin); oils (examples include but are not limited to arachis oil, mineral oil, olive oil, peanut oil, sesame oil and vegetable oil); ointment bases (examples include but are not limited to lanolin, hydrophilic ointment, polyethylene glycol ointment, petrolatum, hydrophilic petrolatum, white ointment, yellow ointment, and rose water ointment); penetration enhancers (transdermal delivery) (examples include but are not limited to monohydroxy or polyhydroxy alcohols, mono-or polyvalent alcohols, saturated or unsaturated fatty alcohols, saturated or unsaturated fatty esters, saturated or unsaturated dicarboxylic acids, essential oils, phosphatidyl derivatives, cephalin, terpenes, amides, ethers, ketones and ureas) plasticizers (examples include but are not limited to diethyl phthalate and glycerol); solvents (examples include but are not limited to ethanol, corn oil, cottonseed oil, glycerol, isopropanol, mineral oil, oleic acid, peanut oil, purified water, water for injection, sterile water for injection and sterile water for irrigation); stiffening agents (examples include but are not limited to cetyl alcohol, cetyl esters wax, microcrystalline wax, paraffin, stearyl alcohol, white wax and yellow wax); suppository bases (examples include but are not limited to cocoa butter and polyethylene glycols (mixtures)); surfactants (examples include but are not limited to benzalkonium chloride, nonoxynol 10, oxtoxynol 9, polysorbate 80, sodium lauryl sulfate and sorbitan mono-palmitate); suspending agents (examples include but are not limited to agar, bentonite, carbomers, carboxymethylcellulose sodium, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, kaolin, methylcellulose, tragacanth and veegum); sweetening agents (examples include but are not limited to aspartame, dextrose, glycerol, mannitol, propylene glycol, saccharin sodium, sorbitol and sucrose); tablet anti-adherents (examples include but are not limited to magnesium 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 opaquants (examples include but are not limited to titanium dioxide); tablet polishing agents (examples include but are not limited to carnuba wax and white wax); thickening agents (examples include but are not limited to beeswax, cetyl alcohol and paraffin); tonicity agents (examples include but are not limited to dextrose and sodium chloride); viscosity increasing agents (examples include but are not limited to alginic acid, bentonite, carbomers, carboxymethylcellulose sodium, methylcellulose, polyvinyl pyrrolidone, sodium alginate and tragacanth); and wetting agents (examples include but are not limited to heptadecaethylene oxycetanol, lecithins, sorbitol monooleate, polyoxyethylene sorbitol monooleate, and polyoxyethylene stearate).

Pharmaceutical compositions according to the present invention can be illustrated as follows:

Sterile IV Solution: A 5 mg/mL solution of the desired compound of this invention can be made using sterile, injectable water, and the pH is adjusted if necessary. The solution is diluted for administration to 1 - 2 mg/mL with sterile 5% dextrose and is administered as an IV infusion over about 60 minutes.

Lvophilized powder for IV administration: A sterile preparation can be prepared with (i) 100 - 1000 mg of the desired compound of this invention as a lypholized 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.

Commercial utility

Component A

Component A

The compounds of formula (I) or pharmaceutically acceptable salts, solvates, hydrates or stereoisomers thereof according to the combination as referred to above are components A. The compounds according to the combination have valuable pharmaceutical properties, which make them commercially utilizable. In particular, they inhibit Bub1 kinase and are expected to be commercially applicable in the therapy of diseases (e.g. cancer).

Component B

Due to the mechanism as discussed above component B is especially suitable to have effects on tumor diseases.

Combination

The combinations of the present invention thus can 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, such as, for example, haematological tumours, solid tumours, and/or metastases thereof.

Such cancer types include, but are not limited, to breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor- resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC) or Non-Small Cell Lung Cancer (NSCLC) and/or metastases thereof.

Additional cancer types, include, but are not limited to, melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof. When the cancer type is cervical cancer, such as cervical adenocarcinoma and/or metastases thereof, component B of the present combinations is preferably docetaxel and component A is preferably compound A1 . Similarly, docetaxel is a preferred component B and compound A1 is a preferred component A in the methods and uses disclosed herein in which the cancer type is cervical cancer such as cervical adenocarcinoma and/or metastases thereof. Preferred uses of the combinations of the invention are for the treatment of Triple- negative breast cancer (TNBC) and/or metastases thereof.

Preferred uses of the combinations of the invention are for the treatment of Castration- Resistant Prostate Cancer (CRPC) and/or metastases thereof.

Preferred uses of the combinations of the invention are for the treatment of NSCLC and/or metastases thereof. Preferred uses of the combinations of the invention are for the treatment of advanced or recurrent TNBC and/or metastases thereof.

Preferred uses of the combinations of the invention are for the treatment of advanced or recurrent CRPC and/or metastases thereof.

Preferred uses of the combinations of the invention are for the treatment of advanced or recurrent NSCLC and/or metastases thereof.

In an embodiment of the present invention the cancer is TNBC and/or metastases thereof.

In an embodiment of the present invention the cancer is CRPC and/or metastases thereof. In an embodiment of the present invention the cancer is NSCLC and/or metastases thereof.

In an embodiment of the present invention the cancer is recurrent NSCLC and/or metastases thereof.

In an embodiment of the present invention the cancer is advanced NSCLC and/or metastases thereof.

In an embodiment of the present invention the cancer is colorectal adenocarcinoma and/or metastases thereof. In an embodiment of the present invention the cancer is glioblastoma/neuroglioma and/or metastases thereof.

In an embodiment of the present invention the cancer is ovarian adenocarcinoma and/or metastases thereof.

In an embodiment of the present invention the cancer is cervical adenocarcinoma and/or metastases thereof. In an embodiment of the present invention the cancer is gastric (stomach) cancer and/or metastases thereof.

In an embodiment of the present invention the cancer is pancreatic carcinoma and/or metastases thereof.

In an embodiment of the present invention the cancer is bladder transitional cell carcinoma and/or metastases thereof.

In an embodiment of the present invention the cancer is NSCLC and/or metastases thereof at any stage as defined in The Revised International System for Staging Lung Cancer, adopted in 2010 by the American Joint Committee on Cancer (AJCC) and the Union Internationale Contre le Cancer (Mountain CF: Revisions in the International System for Staging Lung Cancer. Chest 1 1 1 (6): 1710-7, 1997 ; Lung. In: Edge SB, Byrd DR, Compton CC, et al., eds.: AJCC Cancer Staging Manual. 7th ed. New York, NY: Springer, 2010, pp 253-70., which are included herein by reference in their entirety).

In an embodiment the breast cancer, such as the Triple-negative breast cancer (TNBC), antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, the prostate cancer, such as Castration-Resistant Prostate Cancer (CRPC) or the Non- Small Cell Lung Cancer (NSCLC) and/or metastases thereof is resistant and/or insensitive to treatment with standard of care drugs.

In another embodiment the melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof is resistant and/or insensitive to treatment with standard of care drugs. Techniques and methods to determine whether a cancer is resistant and/or insensitive to treatment with standard of care drugs are readily available to the skilled person, such as the methods provided in the Experimental Section of the present application. It is within the knowledge of the skilled person how to readily adapt/modify such methods to test different tumor models for the same or different indications in order to determine whether a cancer is resistant and/or insensitive to treatment with standard of care drugs. Suitable techniques include in vitro and in vivo methods.

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.

Combinations of the present invention might be utilized to inhibit, block, reduce, decrease, etc., cell proliferation and/or cell division, and/or produce apoptosis.

This invention includes a method comprising administering to a mammal in need thereof, including a human, an amount of a component A or a pharmaceutically acceptable salt, isomer, polymorph, metabolite, hydrate, solvate or ester thereof and an amount of component B of this invention; which is effective to treat a disorder selected from breast cancer, such as Triple-negative breast cancer (TNBC), antiestrogen- resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as Castration- Resistant Prostate Cancer (CRPC) and Non-Small Cell Lung Cancer (NSCLC) and/or metastases thereof.

This invention also includes a method comprising administering to a mammal in need thereof, including a human, an amount of a component A or a pharmaceutically acceptable salt, isomer, polymorph, metabolite, hydrate, solvate or ester thereof and an amount of component B of this invention; which is effective to treat a disorder selected from melanoma, colorectal cancer, such as colorectal adenocarcinoma, glioblastoma/neuroglioma, ovarian cancer, such as ovarian adenocarcinoma, cervical cancer, such as cervical adenocarcinoma, gastric (stomach) cancer, pancreatic cancer, such as pancreatic carcinoma, and bladder cancer, such as bladder transitional cell carcinoma, and/or metastases thereof.

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.

Dose and administration

Component A

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 ingredients 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 ingredients 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 30 mg/kg body weight per day. The total amount of the active ingredients per dose will generally range from about 1 mg to about 500 mg per dose, and preferably from about 20 mg to about 200 mg per dose. Clinically useful dosing schedules of a compound 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.

Component B

The hyper-proliferative, cytotoxic or cytostatic agent taxane, such as docetaxel or paclitaxel, or combinations thereof, can be administered to a patient at a dosage which can range from about 0.1 to about 300 mg/kg of total body weight. Also, the agents can also be administered in conventional amounts routinely used in cancer chemotherapy, particularly in breast cancer, such as TNBC, antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as CRPC and NSCLC and/or metastases thereof.

Typically, 100 - 175 mg/m 2 paclitaxel are administered once every 3 weeks, or alternatively 75-90 mg/m 2 paclitaxel once weekly, the latter being preferred.

Typically, 60 - 100 mg/m 2 docetaxel are administered once every 3 weeks, or alternatively 30-40 mg/m 2 docetaxel once weekly. 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 compounds 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. Suitable dose(s), administration regime(s) and administration route(s) for taxanes, such as docetaxel or paclitaxel; or combinations thereof include those described in the NCCN Clinical Practice Guidelines in Oncology (NCCN guidelines), in particular in the NCCN Guidelines for Breast Cancer 2.2016, in the NCCN Guidelines for Prostate Cancer 2.2017 and in the NCCN Guidelines for NSCLC Version 4.2017 which is included herein by reference in its entirety. Further, suitable dose(s), administration regime(s) and administration route(s) for taxanes, such as docetaxel or paclitaxel; or combinations thereof may be readily determined by standard techniques known to the skilled person. The dose(s), administration regime(s) and administration route(s) may have to be adapted according to, inter alia, the indication, the indication stage, the patient age and/or the patient gender, among other factors. Such adaptations can be readily determined by standard techniques known to the skilled person.

For both the Bub1 inhibitor of general formula (I) and the hyper-proliferative, cytotoxic or cytostatic agent, selected from a taxane, such as docetaxel or paclitaxel, or combinations thereof, the administered dosage of the compound(s) may be modified depending on any superior or unexpected results which may be obtained as routinely determined with this invention. The agents can be administered by any of the conventional routes of administration for these compounds. The preferred route of administration for the hyper- proliferative/cytotoxic/cytostatic agents using this invention is typically by injection which is the same route of administration used for the agent alone. Any of the hyper- proliferative, cytotoxic or cytostatic agents can be administered in combination with an Bub1 inhibitor of general formula (I) by any of the mentioned routes of administration.

For administering the Bub1 inhibitor of general formula (I) and the hyper- proliferative/cytotoxic/cytostatic agent(s), by any of the routes of administration herein discussed, the Bub1 inhibitor of general formula (I) can be administered simultaneously with the hyper-proliferative, cytotoxic or cytostatic agent. This can be performed by administering a single formulation which contains both the Bub1 inhibitor of general formula (I) and the hyper-proliferative/cytotoxic/cytostatic agent or administering the Bub1 inihibitor of general formula (I) compound and the hyperproliferative/cytotoxic/cytostatic agents in independent formulations at the same time to a patient. Alternatively, the Bub1 inhibitor of general formula (I) can be administered in tandem with the hyper-proliferative/cytotoxic/cytostatic agent. The Bub1 inhibitor of general formula (I) can be administered prior to the hyper-proliferative/cytotoxic/cytostatic agent. For example, the Bub1 inihibitor of general formula (I) can be administered once or more times per day up to 28 consecutive days, or once or more times per week up to 4 consecutive weeks followed by administration of the hyper-proliferative, cytotoxic or cytostatic agent. Also, the hyper-proliferative, cytotoxic or cytostatic agent can be administered first followed by adminstration of the Bub1 inihibitor of general formula (I). The choice of sequence administration of the Bub1 inihibitor of general formula (I) relative to the hyper-proliferative/cytotoxic/cytostatic agent may vary for different agents. Also, the hyper-proliferative/cytotoxic or cytostatic agent can be administered using any regimen which is conventionally used for these agents.

In another regimen of administration, the Bub1 inihibitor of general formula (I) and the hyper-proliferative/cytotoxic/cytostatic agent can be administered once or more times per day on the day of administration.

Any of the routes and regimens of administration may be modified depending on any superior or unexpected results which may be obtained as routinely determined with this invention.

Combinations of the present invention

The combinations of the present invention can be used in particular in therapy and prevention, i.e. prophylaxis, of tumour growth and metastases, especially in solid tumours of all indications and stages with or without pre-treatment of the tumour growth, more especially on breast cancer, such as TNBC, antiestrogen-resistant breast cancer and aromatase inhibitor-resistant breast cancer, prostate cancer, such as CRPC or NSCLC and/or metastases thereof, even more preferably on TNBC, CRPC or NSCLC and/or metastases thereof.

Methods of testing for a particular pharmacological or pharmaceutical property are well known to persons skilled in the art.

The combinations of component A and component B of this invention can be administered as the sole pharmaceutical agent or in combination with one or more further pharmaceutical agents C (i.e. component C) where the resulting combination of components A, B and C causes no unacceptable adverse effects. For example, the combinations of components A and B of this invention can be combined with component C, i.e. one or more further pharmaceutical agents, such as known anti- angiogenesis, anti-hyper-proliferative, antiinflammatory, analgesic, immunoregulatory, diuretic, antiarrhytmic, anti-hypercholsterolemia, anti-dyslipidemia, anti-diabetic or antiviral agents, and the like, as well as with admixtures and combinations thereof.

Component C, can be one or more pharmaceutical agents such as 131 1-chTNT, abarelix, abiraterone, aclarubicin, ado-trastuzumab emtansine, afatinib, aflibercept, aldesleukin, alemtuzumab, Alendronic acid, alitretinoin, altretamine, amifostine, aminoglutethimide, Hexyl aminolevulinate,amrubicin, amsacrine, anastrozole, ancestim, anethole dithiolethione, angiotensin II, antithrombin III, aprepitant, arcitumomab, arglabin, arsenic trioxide, asparaginase, axitinib, azacitidine, basiliximab, belotecan, bendamustine, belinostat, bevacizumab, bexarotene, bicalutamide, bisantrene, bleomycin, bortezomib, buserelin, bosutinib, brentuximab vedotin, busulfan, cabazitaxel, cabozantinib, calcium folinate, calcium levofolinate, capecitabine, capromab, carboplatin, carfilzomib, carmofur, carmustine, catumaxomab, celecoxib, celmoleukin, ceritinib, cetuximab, chlorambucil, chlormadinone, chlormethine, cidofovir, cinacalcet, cisplatin, cladribine, clodronic acid, clofarabine, copanlisib , crisantaspase, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, darbepoetin alfa, dabrafenib, dasatinib, daunorubicin, decitabine, degarelix, denileukin diftitox, denosumab, depreotide, deslorelin, dexrazoxane, dibrospidium chloride, dianhydrogalactitol, diclofenac, docetaxel, dolasetron, doxifluridine, doxorubicin, doxorubicin + estrone, dronabinol, eculizumab, edrecolomab, elliptinium acetate, eltrombopag, endostatin, enocitabine, enzalutamide, epirubicin, epitiostanol, epoetin alfa, epoetin beta, epoetin zeta, eptaplatin, eribulin, erlotinib, esomeprazole, estradiol, estramustine, 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, lanreotide, lapatinib, lasocholine, lenalidomide, 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, nedaplatin, nelarabine, neridronic acid, nivolumabpentetreotide, nilotinib, nilutamide, nimorazole, nimotuzumab, nimustine, nitracrine, nivolumab, obinutuzumab, octreotide, ofatumumab, omacetaxine mepesuccinate, omeprazole, ondansetron, oprelvekin, orgotein, orilotimod, oxaliplatin, oxycodone, oxymetholone, ozogamicine, p53 gene therapy, paclitaxel, palifermin, palladium-103 seed, palonosetron, pamidronic acid, panitumumab, pantoprazole, pazopanib, pegaspargase, PEG-epoetin beta (methoxy PEG-epoetin beta), pembrolizumab, pegfilgrastim, peginterferon alfa-2b, pemetrexed, pentazocine, pentostatin, peplomycin, Perflubutane, perfosfamide, Pertuzumab, picibanil, pilocarpine, pirarubicin, pixantrone, plerixafor, plicamycin, poliglusam, polyestradiol phosphate, polyvinylpyrrolidone + sodium hyaluronate, polysaccharide-K, pomalidomide, ponatinib, porfimer sodium, pralatrexate, prednimustine, prednisone, procarbazine, procodazole, propranolol, quinagolide, rabeprazole, racotumomab, radium-223 chloride, radotinib, raloxifene, raltitrexed, ramosetron, ramucirumab, ranimustine, rasburicase, razoxane, refametinib , regorafenib, risedronic acid, rhenium-186 etidronate, rituximab, romidepsin, romiplostim, romurtide, samarium (153Sm) lexidronam, sargramostim, satumomab, secretin, sipuleucel-T, sizofiran, sobuzoxane, sodium glycididazole, sorafenib, stanozolol, streptozocin, sunitinib, talaporfin, 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, tramadol, trastuzumab, trastuzumab emtansine, treosulfan, tretinoin, trifluridine + tipiracil, trilostane, triptorelin, trametinib, trofosfamide, thrombopoietin, tryptophan, ubenimex, valrubicin, vandetanib, vapreotide, vemurafenib, vinblastine, vincristine, vindesine, vinflunine, vinorelbine, vismodegib, vorinostat, vorozole, yttrium-90 glass microspheres, zinostatin, zinostatin stimalamer, zoledronic acid, zorubicin or combinations thereof.

Optional anti-hyper-proliferative agents which can be added as component C to the combination of components A and B of the present invention include but are not limited to compounds listed on the cancer chemotherapy drug regimens in the 1 1 th Edition of the Merck Index, (1996), which is hereby incorporated by reference.

Other anti-hyper-proliferative agents suitable for use as component C with the combination of components A and B of the present 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.

Generally, the use of cytotoxic and/or cytostatic agents as component C in combination with a combination of components A and B of the present invention will serve to: (1 ) yield better efficacy in reducing the growth of a tumor and/or metastasis or even eliminate the tumor and/ or metastasis as compared to administration of either agent alone,

(2) provide for the administration of lesser amounts of the administered chemo- therapeutic agents,

(3) provide for a chemotherapeutic treatment that is well tolerated in the patient with fewer deleterious pharmacological complications than observed with single agent chemotherapies and certain other combined therapies,

(4) provide for treating a broader spectrum of different cancer types in mammals, especially humans,

(5) provide for a higher response rate among treated patients, (6) provide for a longer survival time among treated patients compared to standard chemotherapy treatments,

(8) provide a longer time for tumor progression, and/or

(9) yield efficacy and tolerability results at least as good as those of the agents used alone, compared to known instances where other cancer agent combinations produce antagonistic effects. Without further elaboration, it is believed that one skilled in the art can, using the preceding description, utilize the present invention to its fullest extent. The following preferred specific embodiments are, therefore, to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever.

BHC173007 FC

EXPERIMENTAL SECTION

1. Preparation of components of the present invention

The schemes and procedures described in the art as cited in the present application (see introductory part) disclose general synthetic routes and specific procedures within their experimental sections to arrive at the Bub1 inhibitor compounds which are preferred components A of the present combination.

Compound A1 , 2-{3,5-difluoro-4-[(3-{5-methoxy-4-[(3-methoxypyridin-4- yl)amino]pyrimidin-2-yl}-1 H-indazol-1 -yl)methyl]phenoxy}ethanol can be prepared according to the methods described in WO2016/042084, particularly using the method

Compound A1

Component B is available from commercial sources. Alternatively Component B may be prepared using any of the methods available in the art. 2. Biological in vitro Experiments:

2.1 Test system

Cell line Tumor entity Source Plating cell no

22RV1 prostate carcinoma ATCC CRL-2505 800

NCI-H1299 non-small cell lung carcinoma ATCC CRL-5803 700

Bayer TRG 600 triple-negative mammary

SUM-149 Oncology internal

carcinoma

stock *

A375 malignant melanoma ATCC CRL-1619 600

Colo205 colorectal adenocarcinoma ATCC CCL-222 800

H4 glioblastoma/neuroglioma ATCC HTB-148 700

OVCAR-3 ovarian adenocarcinoma NCI 507262 1 000

HeLa cervical adenocarcinoma ATCC CCL-2 600

Hs746T gastric (stomach) carcinoma ATCC HTB-135 700 triple-negative mammary 800

MDA-MB-436 CLS 300278

carcinoma

MIA Paca-2 pancreatic carcinoma ATCC CRL-1420 600

PANC-1 pancreatic carcinoma ATCC CRL-1469 1200

UM-UC-3 bladder carcinoma ATCC CRL-1749 700

* may also be sourced from commercial vendors, such as Asterand, for example.

2.2 Study design

2.3 Methods and parameters

Tumor cells were propagated in a humidified 37Ό in cubator in their respective growth medium supplemented 10% fetal calf serum. For analysis of combination effects between a compound A and a compound B, cells were plated in 384-well plates at the cell numbers per well as indicated in 2.1 (Study design). After 24h, cells were treated with a Bub1 inhibitor (component A1 ) and with one of the taxanes (component B) paclitaxel or docetaxel for single compound treatments (final concentrations see 2.2 (Study design)), and in nine different fixed-ratio combinations of compound A (D1 ) and compound B (D2) (0.9xD1 +0.1 xD2, 0.8xD1 +0.2xD2, 0.7xD1 +0.3xD2, 0.6xD1 +0.4xD2, 0.5xD1 +0.5xD2, 0.4xD1 +0.6xD2, 0.3xD1 +0.7xD2, 0.2xD1 +0.8xD2, 0.1 xD1 +0.9xD2). Cell viability was assessed after 96 hour exposure with the Cell Titre-Glo Luminescent Cell Viability Assay (Promega). IC50 values (inhibitory concentration at 50% of maximal effect) were determined by means of a 4 parameter fit on measurement data which were normalized to vehicle (DMSO) treated cells (=100%) and measurement readings taken immediately before compound exposure (=0%). IC50 isobolograms were plotted with the actual concentrations of the two compounds on the x- and y-axis, and the combination index (CI) was calculated according to the median-effect model of Chou- Talalay [Chou T.C. Pharmacol. Rev. 58, 621 , 2006]. A CI of <0.8 was defined as more than additive (synergistic) interaction, and a CI of >1 .2 was defined as antagonistic interaction.

2.4 Results

Calculated combination indices (CI50) at IC50 for Bub1 inhibitor plus paclitaxel or docetaxel are summarized in tables 1 and 2, along with the mono-treatment IC50 values and the concentrations required in combination to achieve the CI50.

CI50 interpretation code: CI50 <0.8, synergism; 0.8< CI50≤1 .2, additivity; CI 5 o>1.2, antagonism.

2.4.1 Combination of compound A1 with paclitaxel

Table 1 summarizes the data for Compound A1 plus paclitaxel.

Table 1 : Calculated combination indices (CI) from proliferation assays of cell lines treated with combinations of Bub1 inhibitor Compound A1 and paclitaxel.

Mono-treatment IC50 values and the concentrations required in combination of the two test compounds to achieve the CI50 are shown. All concentrations are given in mol/L.

2.4.2 Combination of compound A1 with docetaxel

Table 2 summarizes the data for Compound A1 plus docetaxel. Table 2: Calculated combination indices (CI) from proliferation assays of cell lines treated with combinations of Bub1 inhibitor Compound A1 and docetaxel.

Mono-treatment IC50 values and the concentrations required in combination of the two test compounds to achieve the CI50 are shown. All concentrations are given in mol/L.

3. Biological in vivo Experiments:

3.1 Combination of compound A1 with paclitaxel in a triple-negative breast cancer model

3.1.1 Test system, study design and methods

Experiments were initiated after an acclimatization period of at least 7 days. Animals were kept in a 12 hours light/dark cycle, food and water was available ad libitum and housing temperature was 23 Ό. All animal experimen ts were conducted in accordance with the German animal welfare law and approved by local authorities.

Study design

Animals were randomly assigned to experimental groups, twelve animals per group. At the initiation of the treatment, animals were tattooed and each cage was labeled with the cage number, study number and number of animals per cage.

Animals and strain Female, 5-6 weeks old NM RI nude mice (BomTac:NM RI-Foxn1 nu), body weight 20-22 g

Supplier Taconic M&B A/S (Denmark)

Tumor transplantation a single s. c. injection of 5 x 1 0 s SUM-149 tumor cells suspended in

0.1 ml Matrigel

Start of treatment treatment was started when the tumor area reached a

predetermined size in mice of 25 to 30 mm 2

Randomization tumor bearing mice were randomized according to the tumor area

Appl volume and route of 0.1 ml / 10 g body weight via a gastric tube p.o. or via injection i.v. administration

Vehicle 1 (Compound A1 ) PEG400 90%, Ethanol 1 0%

Vehicle 2 (Paclitaxel) Cremophor 5% / Ethanol 5% / Saline 90%

Treatment Schedule

Treatment groups No of Dose Drug cone. Schedule of animals [mg/kg] [mg/mL vehicle] administrations, route

1 . Vehicle control 12 -- vehicle 1 2QD, p.o.

2. Compound A1 12 25 2.5 2QD, p.o.

3. Paclitaxel 12 20 2.0 1 QW, i.v.

4. Compound A1 12 25 2.5 2QD, p.o.

+ Paclitaxel 20 2.0 1 QW, i.v.

Methods and parameters

SUM-149 human breast cancer cells from Bayer-internal cell stock were cultured in DMEM/Ham ' s F12 medium containing stable glutamine (Biochrom, Germany) supplemented with 5% fetal calf serum (Biochrom, Germany), 1 μg/ml hydrocortisone (Biochrome, Germany), and 5 Mg/ml bovine insulin (Biochrome, Germany). Cells were harvested for transplantation in a subconfluent (70%) state. Animals were injected with 5 x 10 6 SUM-149 cells suspended in 100% Matrigel into the left inguinal region on day 0.

When tumors reached a predetermined size in mice of 25-30 mm 2 animals were randomized into treatment and control groups (n=12 animals/group) and treatment with Compound A1 , Paclitaxel, or combination started. The oral application volumes were 10 ml/kg for mice, the intravenous application volume was 10 ml/kg for mice. The time interval between two applications per day was 6-7h.

Tumor response was assessed by determination of the tumor area (product of the longest diameter and its perpendicular) using a caliper. The animal body weight was monitored as a measure for treatment-related toxicity. Measurement of tumor area and body weight were performed three times weekly.

Animals were sacrificed when showing signs of toxicity (>20% body weight loss) or when tumors reached a size of approximately 120 mm 2 .

Tumor growth inhibition is presented as T/C ratio (Treatment / Control) calculated with tumor areas when the vehicle control group had to be closed. Relative tumor growth inhibition based on tumor area (relative T/C) was calculated by the formula [(tumor area of treatment group at day x) - (tumor area of treatment group at day before first treatment)] / [(tumor area of vehicle group at day x) - (tumor area of vehicle group at day before first treatment)].

Statistical analysis

Statistical analysis was assessed using SigmaStat software. A Mann-Whitney Rank Sum test and Analysis of Variance (ANOVA) on ranks were performed. When P values <0.05 it is designated statistically significant difference.

3.1.2 Results

Treatment for all groups started at a tumor size of approx. 27 mm 2 , at day 10 after tumor cell inoculation. Groups 1 and 2 were treated until day 38 when they had to be terminated for animal welfare reasons whereas treatment of the other groups was continued. Efficacy of treatment of groups 3 and 4 was evaluated on day 54 (Table 3, Figure 5).

At day 38 when the vehicle group had to be terminated due to tumor size, Compound A1 (group 2) showed only marginal single agent efficacy (relative T/Carea 0.84), whereas Paclitaxel (group 3) was efficacious (relative T/Carea 0.17). Surprisingly, the combination of Compound A1 and Paclitaxel (group 4) was highly efficacious (relative T/Carea 0.02). The mean tumor areas of treatment groups 3 and 4 were statistically different from the vehicle group (p<0.05). Paclitaxel-treated tumors showed a continuous growth even under continued Paclitaxel treatment and reached a size of approximately 99 mm 2 at day 54. Surprisingly, the animals treated with the combination of Compound A1 and Paclitaxel showed a strongly reduced tumor growth rate and mean relative tumor area at day 54 was 70% below the mean relative tumor area of the Paclitaxel treated tumors. The difference in tumor areas between the Paclitaxel single agent treatment group and the combination treatment group were statistically significant (P<0.001 ). Table 3: Time course of SUM-149 triple-negative (TN) breast cancer xenograft tumor growth.

Tumor area in mm 2 measured at the respective days after tumor inoculation. Treatment with vehicle (control), Compound A1 , paclitaxel, or the combination of Compound A1 and paclitaxel started on day 10. Compound A1 was administered twice daily (2QD) p.o. at a dose of 25 mg/kg, paclitaxel was administered once daily i.v. 1 day on/ 6 days off (QW) at a dose of 20 mg/kg.

3.2 Combination of compound A1 with paclitaxel in a non-small cell lung cancer model

3.2.1 Test system, study design and methods

Experiments were initiated after an acclimatization period of at least 7 days. Animals were kept in a 12 hours light/dark cycle, food and water was available ad libitum and housing temperature was 23 Ό. All animal experimen ts were conducted in accordance with the German animal welfare law and approved by local authorities. Study design

Animals were randomly assigned to experimental groups, twelve animals per group. At the initiation of the treatment, animals were tattooed and each cage was labeled with the cage number, study number and number of animals per cage.

Methods and parameters

NCI-H1299 human non-small cell lung cancer cells originally purchased from ATCC (Manassas, VA, USA; #CRL-5803) were cultured as described according to the supplier ' s protocols. Cells were harvested for transplantation in a subconfluent (70%) state. Animals were injected with 3 x 10 6 NCI-H1299 cells suspended in 100% Matrigel into the left inguinal region on day 0.

When tumors reached a predetermined size in mice of 12-22 mm 2 animals were randomized into treatment and control groups (n=12 animals/group) and treatment with Compound A1 , Paclitaxel, or combination started. The oral application volumes were 10 ml/kg for mice, the intravenous application volume was 10 ml/kg for mice. The time interval between two applications per day was 6-7h. Tumor response was assessed by determination of the tumor area (product of the longest diameter and its perpendicular) using a caliper. The animal body weight was monitored as a measure for treatment-related toxicity. Measurement of tumor area and body weight were performed three times weekly.

Animals were sacrificed when showing signs of toxicity (>20% body weight loss) or when tumors reached a size of approximately 120 mm 2 .

Tumor growth inhibition is presented as T/C ratio (Treatment / Control) calculated with tumor areas when the vehicle control group had to be closed. Relative tumor growth inhibition based on tumor area (relative T/C) was calculated by the formula [(tumor area of treatment group at day x) - (tumor area of treatment group at day before first treatment)] / [(tumor area of vehicle group at day x) - (tumor area of vehicle group at day before first treatment)].

Statistical analysis

Statistical analysis was assessed using SigmaStat software. A T-test and a One Way Analysis of Variance (ANOVA) were performed. When P values <0.05 it is designated statistically significant difference.

3.2.2 Results Treatment for all groups started at a tumor size of approx. 18 mm 2 , at day 7 after tumor cell inoculation. Groups 1 and 2 were treated until day 33 when they had to be terminated for animal welfare reasons whereas treatment of the other groups was continued. Efficacy of treatment of groups 3 and 4 was evaluated on day 54 when necrosis of some xenograft tumors in both groups became emerging (Table 4, Figure 6).

At day 33 when the vehicle group had to be terminated due to tumor size, Compound A1 (group 2) showed only marginal single agent efficacy (relative T/Carea 0.84), whereas Paclitaxel (group 3) was highly efficacious (relative T/C are a 0.1 1 ). Surprisingly, the combination of Compound A1 and Paclitaxel (group 4) was even more efficacious (relative T/Carea 0.04). The mean tumor areas of treatment groups 3 and 4 were statistically different from the vehicle group (p<0.05). Paclitaxel-treated tumors showed a continuous growth even under continued Paclitaxel treatment and reached a size of approximately 93 mm 2 . Surprisingly, the animals treated with the combination of Compound A1 and Paclitaxel showed a strongly reduced tumor growth rate and mean relative tumor area at day 54 was 70% below the mean relative tumor area of the Paclitaxel treated tumors. The difference in tumor areas between the Paclitaxel single agent treatment group and the combination treatment group were statistically significant (P<0.05).

5

Table 4: Time corse NCI-H1299 non-small cell lung cancer (NSCLC) xenograft tumor growth.

Tumor area in mm 2 measured at the respective days after tumor inoculation. Treatment with vehicle (control), Compound A1 , paclitaxel, or the combination of Compound A1 10 and paclitaxel started on day 7. Compound A1 was administered twice daily (2QD) p.o. at a dose of 50 mg/kg in the single agent treatment group and at 25 mg/kg in combination with paclitaxel, paclitaxel was administered once daily i.v. 1 day on/ 6 days off (QW) at a dose of 20 mg/kg (days 7-39) and 15 mg/kg (days 40-54).

15

3.3 Combination of compound A1 with paclitaxel in a castration-resistant prostate cancer model

3.3.1 Test system, study design and methods

20

Experiments were initiated after an acclimatization period of at least 7 days. Animals were kept in a 12 hours light/dark cycle, food and water was available ad libitum and housing temperature was 23 Ό. All animal experimen ts were conducted in accordance with the German animal welfare law and approved by local authorities.

25 Study design

Animals were randomly assigned to experimental groups, twelve animals per group. At the initiation of the treatment, animals were tattooed and each cage was labeled with the cage number, study number and number of animals per cage.

Methods and parameters

22Rv1 human castration-resistant prostate cancer cells originally purchased from ATCC (Manassas, VA, USA; #CRL-2505) were cultured as described according to the supplier ' s protocols. Cells were harvested for transplantation in a subconfluent (70%) state. Animals were injected with 3 x 10 6 22Rv1 cells suspended in 100% Matrigel into the left inguinal region on day 0.

When tumors reached a predetermined size in mice of approximately 41 mm 2 animals were randomized into treatment and control groups (n=12 animals/group) and treatment with Docetaxel or the combination of Compound A1 and Docetaxel started. The oral application volumes were 10 ml/kg for mice, the intravenous application volume was 10 ml/kg for mice. The time interval between two applications per day was 6-7h. Tumor response was assessed by determination of the tumor area (product of the longest diameter and its perpendicular) using a caliper. The animal body weight was monitored as a measure for treatment-related toxicity. Measurement of tumor area and body weight were performed three times weekly.

Animals were sacrificed when showing signs of toxicity (>20% body weight loss) or when tumors reached a size of approximately 225 mm 2 .

Tumor growth inhibition is presented as T/C ratio (Treatment / Control) calculated with tumor areas when the vehicle control group had to be closed. Relative tumor growth inhibition based on tumor area (relative T/C) was calculated by the formula [(tumor area of treatment group at day x) - (tumor area of treatment group at day before first treatment)] / [(tumor area of vehicle group at day x) - (tumor area of vehicle group at day before first treatment)].

Statistical analysis

Statistical analysis was assessed using SigmaStat software. A T-test and a One Way Analysis of Variance (ANOVA) were performed. When P values <0.05 it is designated statistically significant difference.

3.3.2 Results Treatment for all groups started at a tumor size of approx. 42 mm 2 , at day 12 after tumor cell inoculation. All groups were treated until day 27 when they had to be terminated for animal welfare reasons (Table 5, Figure 7).

Treatment with Docetaxel (group 2) showed only marginal single agent efficacy (relative T/Carea 0.75). Surprisingly, the combination of Compound A1 and Docetaxel (group 3) was much more efficacious (relative T/C are a 0.52) and mean relative tumor area at day 27 was 48% below the mean relative tumor area of the Docetaxel treated tumors. The mean tumor areas of all treatment groups were statistically different from the vehicle group (p<0.05). The difference in tumor areas between the Docetaxel single agent treatment group and the combination treatment group were statistically significant (P=0.017). Table 5: Time course of 22Rv1 castration-resistant prostate cancer (CRPC) xenograft tumor growth.

Tumor area in mm 2 measured at the respective days after tumor inoculation. Treatment with vehicle (control), docetaxel, or the combination of Compound A1 and docetaxel started day 12. Compound A1 was administered twice daily (2QD) p.o. at a dose of 50 mg/kg, docetaxel was administered once daily i.v. 1 day on/ 6 days off (QW) at a dose of 4 mg/kg.

4. Conclusions:

In summary, in the present studies the combination of Bub1 kinase inhibitor Compound A1 with the taxanes Paclitaxel and Docetaxel in proliferation assays of human carcinoma cells showed the utility of the present invention. The in vitro results showed predominantly more than additive (CI 5 o <0.8) and in one cell line (SUM-149) additive (0.8< Clso≤1.2) interaction.

The in vivo xenograft studies in the SUM-149 triple-negative human breast cancer model, the NCI-H1299 non-small cell lung cancer model, and the 22Rv1 castration- resistant prostate model in nude or SCID mice clearly demonstrated the more than additive anti-tumor efficacy of the combination of a Bub1 kinase inhibitor with a taxane (herein shown with Paclitaxel or Docetaxel) as compared to the respective single agent efficacies. Surprisingly, combining an Bub1 kinase inhibitor with a taxane strongly deminished the outgrowth of the tumors under continued treatment as compared to taxane single agent treatment. Most surprising was the strong more than additive in vivo efficacy in the SUM-149 tumor model which could not be predicted from the additive interaction observed in vitro for the combination of a Bub1 inhibitor with paclitaxel.

Also advantageously, the combination of Bub1 kinase inhibitor Compound A1 with taxanes did not show increased toxicity over taxane monotherapy.

These results demonstrate that combination of Bub1 kinase inhibitors with taxanes can result in more than additive anti-proliferative (synergistic) efficacy in tumor cells and in vivo tumor models and warrant further clinical evaluation of this promising combination therapy for the treatment of cancer.

Description of the Figures

Figure 1

Compound A1 plus paclitaxel in vitro combination assay with SUM-149 cells. SUM-149 human triple-negative breast cancer cells were grown in the presence various concentrations of Compound A1 (0.1 -10 μΜ) and paclitaxel (1 -100 nM) in mono and in nine different fixed-ratio combinations. IC50 values were determined and the respective Compound A1 and paclitaxel concentrations plotted in IC50 Isobolograms. The grey dashed lines indicate the results expected for additivity.

Figure 2

Compound A1 plus paclitaxel in vitro combination assay with NCI-H1299 cells. NCI- H1299 human non-small cell lung cancer cells were grown in the presence various concentrations of Compound A1 (0.1 -10 μΜ) and paclitaxel (1 -100 nM) in mono and in nine different fixed-ratio combinations. IC50 values were determined and the respective Compound A1 and paclitaxel concentrations plotted in IC50 Isobolograms. The grey dashed lines indicate the results expected for additivity.

Figure 3

Compound A1 plus docetaxel in vitro combination assay with NCI-H1299 cells. NCI- HI 299 human non-small cell lung cancer cells were grown in the presence various concentrations of Compound A1 (0.1 -10 μΜ) and docetaxel (0.3-30 nM) in mono and in nine different fixed-ratio combinations. IC50 values were determined and the respective Compound A1 and paclitaxel concentrations plotted in IC50 Isobolograms. The grey dashed lines indicate the results expected for additivity.

Figure 4

Compound A1 plus docetaxel in vitro combination assay with 22Rv1 cells. 22Rv1 human prostate cancer cells were grown in the presence various concentrations of Compound A1 (0.1 -10 μΜ) and docetaxel (0.3-30 nM) in mono and in nine different fixed-ratio combinations. IC50 values were determined and the respective Compound A1 and paclitaxel concentrations plotted in IC50 Isobolograms. The grey dashed lines indicate the results expected for additivity.

Figure 5 Time course of SUM-149 triple-negative (TN) breast cancer xenograft model on female nude mice and body weight change (%). Treatment with Compound A1 , paclitaxel, or the combination of Compound A1 and paclitaxel started on day 10. Compound A1 was administered twice daily (2QD) p.o. at a dose of 25 mg/kg, paclitaxel was administered once daily i.v. 1 day on/ 6 days off (QW) at a dose of 20 mg/kg.

Figure 6

Time course of NCI-H1299 non-small cell lung cancer (NSCLC) xenograft model on female nude mice and body weight change (%). Treatment with Compound A1 , paclitaxel, or the combination of Compound A1 and paclitaxel started on day 7. Compound A1 was administered twice daily (2QD) p.o. at a dose of 50 mg/kg in the single agent treatment group and at 25 mg/kg in combination with paclitaxel, paclitaxel was administered once daily i.v. 1 day on/ 6 days off (QW) at a dose of 20 mg/kg (days 7 -39) and 15 mg/kg (days 40-54).

Figure 7

Time course of 22Rv1 castration-resistant prostate cancer (CRPC) xenograft model on male SCID mice and body weight change (%). Treatment with docetaxel or the combination of Compound A1 and docetaxel started on day 12. Compound A1 was administered twice daily (2QD) p.o. at a dose of 50 mg/kg, docetaxel was administered once daily i.v. 1 day on/ 6 days off (QW) at a dose of 4 mg/kg.