Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SUBSTITUTED PYRAZOLOPYRIDINAMINES
Document Type and Number:
WIPO Patent Application WO/2017/081003
Kind Code:
A1
Abstract:
The present invention relates to substituted pyrazolopyridinamine compounds of general formula (I) as described and defined herein, to methods of preparing said compounds, to intermediate compounds useful for preparing said compounds, to pharmaceutical compositions and combinations comprising said compounds and to the use of said compounds for manufacturing a pharmaceutical composition for the treatment or prophylaxis of a disease, in particular of a hyperproliferative, angiogenesis disorders, inflammatory diseases or diseases associated with inflammatory pain, as a sole agent or in combination with other active ingredients.

Inventors:
KLAR ULRICH (DE)
SCHMIDT NICOLE (US)
GIESE ANJA (DE)
KETTSCHAU GEORG (DE)
MENZ STEPHAN (DE)
SCHÄCKE HEIKE (DE)
RAUSCH ALEXANDRA (DE)
HAUFF PETER (DE)
GRÖTICKE INA (DE)
Application Number:
PCT/EP2016/076978
Publication Date:
May 18, 2017
Filing Date:
November 08, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BAYER PHARMA AG (DE)
International Classes:
C07D519/00; A61K31/519; A61P35/00
Domestic Patent References:
WO2016096721A12016-06-23
WO2013174744A12013-11-28
Attorney, Agent or Firm:
BIP PATENTS (DE)
Download PDF:
Claims:
CLAIMS

1. A compound of general formula (I) :

in which :

R1 represents a halogen atom or a group selected from:

hydroxy-, cyano-, Ci-Ce-alkyl-, d-Ce-alkoxy-, halo-Ci-Ce-alkyl-, halo-d-Ce-alkoxy-, hydroxy-Ci-Ce-alkyk Ci-C3-alkoxy-Ci-C3-alkyl-, S-(CrC3-alkyl)

R2 represents a hydrogen atom or a group Ci-Ce-alkyl-;

R3 represents a group selected from: -C(=0)-0-R5, -C(=0)-N(R5a)R5;

R4 represents halo-, hydroxy-, oxo- (0=), cyano-, nitro-, Ci-Ce-alkyl-,

C2-Ce-alkenyl-, d-Ce-alkynyl-, halo-Ci-Ce-alkyl-, Ci-Ce-alkoxy-,

halo-Ci-Ce-alkoxy-, hydroxy-Ci-Ce-alkyl-, Ci-Ce-alkoxy-Ci-Ce-alkyl-, halo-Ci-Ce-alkoxy-Ci-Ce-alkyl-, Re-0-, -C(=0)-Re, -C(=0)-0-R6, -O- C(=0)-R6, -N(R6a)-C(=0)-R6b, -N(R6a)-C(=0)-0-R6b, -N(R6a)-C(=0)-N(R6b)R6c, - N(R6a)R6b, -N(R6a)R6d, -C(=0)-N(R6a)R6b, Re-S-, R6-S(=0)-, Re-S(=0)2-, -N(R6a)- S(=0)-R6b, -S(=0)-N(R6a)R6b, -N(Res)-S(=0)2-

R6b, -S(=0)2-N(R6a)R6b, -S(=0)=N(R6a)R6b, -N=S(=0)(R6a)R6b or -(Ci-Ce-alkyl)-

N(R6a)R6b;

R5, R5a, R5b are the same or different and independently selected from each other, and represent a hydrogen atom or a group selected from: Ci-Ce-alkyl-, Cs-Ce-cycloalkyl-, 3- to 10-membered heterocycloalkyi-, aryl-, and heteroaryl-; wherein said Ci-Ce-alkyl-, d-Ce-cycloalkyk 3- to 10-membered

heterocycloalkyi-, aryl- and heteroaryl- group is optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R4 groups ;or

N(R5a)R5 together represent a 3- to 10-membered heterocycloalkyi- group, said group being optionally substituted, identically or differently, with 1 , 2, 3. 4 or 5 R4 groups ; R6, R6a, R6b, R6care the same or different and independently selected from each other, and represent a hydrogen atom, a d-Ce-alkyl- or a C3-Ce-cycloalkyl- group: or

R6a and R6b, or

R6a a n d R6C j or

R6b and R6c together may form a C2-Ce-alkylene group, in which optionally one

methylene can be replaced by -0-, -C(=0)-, -NH-, or -N(Ci-C4-alkyl)- ;

R6d represents -(C -C6-alkyl)-N(Rea)R6b ;

or a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same

2. The compound according to claim 1 , wherein

R1 represents a halogen atom or a group selected from: Ci-C3-alkyk Ci-C3-alkoxy-, halo-Ci-Cr-alkyl-, and -S-(Ci-C3-alkyl).

3. The compound according to claim 1 or 2, wherein

R represents a chlorine atom or a -O-CH3 group.

4. The compound according to any one of claims 1 to 3, wherein

R2 represents a hydrogen atom.

5. The compound according to any one of claims 1 to 4, wherein

R3 represents a group -C(=0)-N(R5a)R5.

6. The compound according to any one of claims 1 to 5, wherein

R4 represents halo-, hydroxy-, cyano-, C -C3-alkyl-, Ci-C3-alkoxy-, -N(R6a)-C(=0)-0-R6i -N(R6a)R6b, -N(R6a)R6d, -C(=0)-N(Rea)R6b or -(Ci-C3-alkyl)-N(R6a)R6b.

7. The compound according to any one of claims 1 to 6, wherein

R5a, R5b are the same or different and are independently selected from R5 ;

R5 represents a hydrogen atom or a C -C3-alkyl- group:

wherein said C -C3-alkyl- group is optionally substituted, identically or differently. with 1 , 2 or 3 R4 groups.

8. The compound according to any one of claims 1 to 7, wherein

N(R5a)R5 together represent a 3- to 10-membered heterocycloalkyi- group, said group being optionally substituted, identically or differently, with 1 or 2 R4 groups.

9. The compound according to any one of claims 1 to 8. wherein

6a, R6b, R6c are the same or different and are independently selected from R6 ;

R6 represents a hydrogen atom or a Ci-Cs-alkyl- group: and

R6d represents a -(Ci-C3-alkyl)-N(R6a)R6b group.

10. The compound according to claim 1 , which is selected from the group consisting of :

(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-N,N-dimethyl-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide,

(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl[3-(dimethylamino)azetidin-1- yl]methanone.

(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-N-(2-methoxyethyl)-N-methyl- 5,6J,8-tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide,

(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yi)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl[(3R)-3-(dimethylamino)pyrrolidin-1- yljmethanone,

(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7!8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl[(2R,6S)-2,6-dimethylmorpholin-4- yljmethanone,

(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)aminoj-5,6,7,8- tetrahydro[1]benzothieno[2.3-d]pyrimidin-7-yl4-[2-(dimethylamino)ethyl]piperazin- 1 -ylmethanone.

(7S)-4-[(4-methoxypyrazolo[1 ^-ajpyridin-S-y aminoj-N^-dimethyl-S.GJ.S- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide,

[3-(dimethylamino)azetidin-1-yl](7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)aminoj- 5,6J,8-tetrahydro[1]benzothieno[2.3-d]pyrimidin-7-ylmethanone,

(7S)-N-(2-methoxyethyl)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-N-methyl- 5,6J,8-tetrahydro[1]benzothieno[2.3-d]pyrimidine-7-carboxamide,

[(3R)-3-(dimethylamino)pyrrolidin-1-yl](7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5- yl)amino]-5,6J,8-tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-ylmethanone,

[(2R,6S)-2,6-dimethylmorpholin-4-yl](7S)-4-[(4-methoxypyrazolo[1 !5-a]pyridin-5- yl)amino]-5,6,7,8-tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-ylmethanone, 4-[2-(dimeihylamino)eihyl]piperazin-1-yl(7S)-4-[(4-meihoxypyrazolo[1 ,5-a]pyridin-5- yl)amino]-5,6J,8-tetra ydro[1]benzot ieno[2,3-d]pyrimidin-7-ylmet anone,

(4 [2-(dimethylamino)ethyl](meihyl)amino}piperidin-1-yl){(7S)-4-[(4- met oxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8-tetrahydro[1]benzoihieno[2,3- d]pyrimidin-7-yl}methanone,

{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}(4-methylpiperazin-1 -yl)methanone,

(3-hydroxy-3-methylazetidin-1-yl){(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]- 5,6J,8-tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}methanone,

(7S)-N-[2-(dimethylamino)ethyl]-4-[(4-methoxypyrazolo[1 (5-a]pyridin-5-yl)amino]-

5!6,7,8-tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide,

{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6J,8- ietrahydro[1]benzothieno[2!3-d]pyrimidin-7-yi}[(1 S,4S)-5-meihyl-2,5- diazabicyclo[2.2.1 ]hept-2-yl]methanone,

{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- ieirahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(1 R,4 )-5-meihyl-2,5- diazabicyclo[2.2.1 ]hept-2-yl]methanone.

{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2.3-d]pyrimidin-7-yl}[(1 S,4S)-2-oxa-5- azabicyclo[2.2.1 ]hept-5-yl]methanone.

{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(1 R,4R)-2-oxa-5- azabicyc!o[2.2.1 ]hept-5-yl]methanone,

[4-(dimeihylamino)piperidin-1-yl]{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]- 5,6,7,8-ietrahydro[13benzothieno[2,3-d]pyrimidin-7-yl}methanone,

{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(3S)-3-methylmorpholin-4- yl]methanone.

{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(3R)-3-methylmorpholin-4- yljmethanone,

{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}(morpholin-4-yl)methanone, [(3S)-3-(dimethylamino)pyrrolidin-1-yl]{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5- yl)amino]-5,6J,8-tetra ydro[1]benzot ieno[2,3-d]pyrimidin-7-yl}methanone,

1 -({(7S)-4-[(4-methoxypyrazo!o[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- ietrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}carbonyl)azetidine-3-carbonitrile,

(7S)-N-[3-(dimethylamino)propyl]-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-N- meihyl-5,6,7,8-tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide,

(7S)-N-[2-(dimethylamino)ethyl]-N-ethyl-4-[(4-meihoxypyrazolo[1 ,5-a]pyridin-5- yl)amino]-5!6!7,8-ietrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide,

(7S)-N-[2-(dimethylamino)ethyl]-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-N- methyl-5,6,7,8-ieirahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide,

(7S)-N-(2-hydroxyeihyl)-4-[(4-meihoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-N-methyl- 5,6,7, 8-ietrahydro[1]benzoihieno[2,3-d]pyrimidine-7-carboxamide,

(7S)-N-(2,2-difiuoroethyl)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-N-methyl-

5,6,7,8-leirahydro[1]benzoihieno[2,3-d]pyrimidine-7-carboxamide,

(7S)-N-(2-meihoxyethyl)-4-[(4-meihoxypyrazo!o[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide,

(7S)-N-isopropyl-4-[(4-meihoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide,

(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-N-methyl-5,6,7,8- tetrahydro[1]benzothieno[2.3-d]pyrimidine-7-carboxamide.

(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-N-isopropyl-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide,

(7S)-4-[(4-chloropyrazo!o[1 ,5-a]pyridin-5-yl)amino]-N-ethyl-N-methyl-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide,

(7S)-4-[(4-chloropyrazo!o[1 ,5-a]pyridin-5-yl)amino]-N-(2-hydroxyeihyl)-N-melhyl-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide,

(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-N-[2-(dimethylamino)ethyl]-N-ethyl- 5,6.7,8-tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide,

1 -({(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}carbonyl)azetidine-3-carbonitrile, {(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetra ydro[1]benzothieno[2,3-d]pyrimidin-7-yl}(3-hydroxy-3-met ylazetidin-1 - yl)methanone.

{(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[4-(dimethylamino)piperidin-1- yljmethanone,

{(7S)-4-[(4-chloropyrazolo[1.5-a]pyridin-5-yl)amino]-5, 6,7-8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}(4-{[2- (dimethylamino)ethyl](methyl)amino}piperidin-1 -yl)methanone,

{(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5!6!7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}(4-methylpiperazin-1 -yl)methanone,

{(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yi}[(1 S.4S)-2-oxa-5- azabicyclo[2.2.1 ]hept-5-yl]methanone.

{(7S)-4-[(4-chloropyrazolo[1.5-a]pyridin-5-yl)amino]-5,6,7!8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(1 ,4 )-2-oxa-5- azabicyclo[2.2.1 ]hept-5-yl]methanone.

{(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydrotljbenzothieno^^-djpyrimidin^-ylJKI S^SVS-methyl^^- diazabicyclo[2.2.1 ]hept-2-yl]methanone,

{(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(1 ,4R)-5-methyl-2,5- diazabicyclo[2.2.1 ]hept-2-yl]methanone,

{(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}(4-{[2- (dimethylamino)ethyl](methyl)amino}piperidin-1 -yl)methanone,

{(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(3S)-3-(dimethylamino)pyrrolidin-1- yljmethanone

tert-butyl [1 -({(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}carbonyl)azetidin-3-yl]carbamate

(3-aminoazetidin-1 -yl){(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}nnethanone (3-aminoazeiidin-1 -yl){(7S)-4-[(4-meihoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2.3-d]pyrimidin-7-yl}methanone dihydrochloride

((7S)-4-[(4-methoxypyrazolo[1.5-a]pyridin-5-yl)amino]-N-

(pentacyclo[4.2.0.02,5.03,8.04,7]oct-1 -yl)-5!6!7,8-tetrahydro[1]benzothieno[2, 3- d]pyrimidine-7-carboxamide

tert-butyl [1 -({(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7.8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}carbonyl)azetidin-3-yl]carbamate

(3-aminoazeiidin-1 -yl){(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6J,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}methanone

[(3R)-3,4-dimeihylpiperazin-1-yl]{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-

5,6,7,8-teirahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}methanone

[(3S)-3.4-dimethylpiperazin-1-yl]{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]- 5,6,7,8-tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}methanone

{(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)annino]-5,6,7,8- ieirahydro[1]benzoihieno[2,3-d]pyrimidin-7-yl}[(3R)-3,4-dimethylpiperazin-1- yljmethanone

{(7S)-4-[(4-chloropyrazolo[1 !5-a]pyridin-5-yl)amino]-5,6,7!8- teirahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(3S)-3,4-dimeihylpiperazin-1- yljmethanone

or a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.

11. A method of preparing a compound of general formula (I) according to any one of

claims 1 to 10, in which method ompound of general formula (II) :

in which R2 and R3 are as defined for general formula (I) in any one of claims 1 to 13, and LG represents a leaving group, preferably a chlorine atom;

is allowed to react with an intermediate compound of general formula (III):

(I")

in which R1 is as defined for general formula (I) in any one of claims 1 to 13.

12. A compound of general formula (I), or a stereoisomer, a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, according to any one of claims 1 to 10, for use in the treatment or prophylaxis of a disease.

13. A pharmaceutical composition comprising a compound of general formula (I), or a stereoisomer, a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, according to any one of claims 1 to 10, and a pharmaceutically acceptable diluent or carrier.

14. A pharmaceutical combination comprising :

one or more first active ingredients selected from a compound of general formula (I) according to any of claims 1 to 10, and

one or more second active ingredients selected from chemotherapeutic anti-cancer agents.

15. Use of a compound of general formula (I), or a stereoisomer, a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, according to any one of claims 1 to 10, for the prophylaxis or treatment of a disease.

16. Use of a compound of general formula (I), or a stereoisomer, a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, according to any one of claims 1 to 10, for the preparation of a medicament for the prophylaxis or treatment of a disease.

17. Use according to claims 12, 15 or 16, wherein said disease is a disease of uncontrolled cell growth, proliferation and/or survival, an inappropriate cellular immune response, or an inappropriate cellular inflammatory response, particularly in which the uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune response, or inappropriate cellular inflammatory resnonse is mediated by the MKNK1 pathway, more particularly in which the disease of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune response, or inappropriate cellular inflammatory response is a haematological tumour, a solid tumour and/or metastases thereof, e.g. leukaemias and myelodysplastic syndrome, malignant lymphomas, head and neck tumours including brain tumours and brain metastases, tumours of the thorax including non-small cell and small cell lung tumours, gastrointestinal tumours, endocrine tumours, mammary and other gynaecological tumours, urological tumours including renal, bladder and prostate tumours, skin tumours, and sarcomas, and/or metastases thereof, or pancreatitis.

18. Use according to claims 12, 15 or 16, wherein said disease is one of the following

inflammatory and/or pain-associated diseases

19. Use according to claims 12, 15 or 16, for the treatment of pain syndromes including acute, chronic, inflammatory and neuropathic pain, preferably inflammatory pain, surgical pain, visceral pain, dental pain, premenstrual pain, endometriosis-associated pain, or other endometriosis-associated symptoms, wherein said symptoms are in particular endometriosis-associated dysmenorrhea, dyspareunia, dysuria, or dyschezia, pain associated with fibrotic diseases, central pain, pain due to burning mouth syndrome, pain due to burns, pain due to migraine, cluster headaches, pain due to nerve injury, pain due to neuritis, neuralgias, pain due to poisoning, pain due to ischemic injury, pain due to interstitial cystitis, cancer pain, pain due to viral, parasitic or bacterial infections, pain due to traumatic nerve-injury, pain due to post-traumatic injuries (including fractures and sport injuries), pain due to trigeminal neuralgia, pain associated with small fiber neuropathy, pain associated with diabetic neuropathy, chronic lower back pain, phantom limb pain, pelvic pain syndrome, chronic pelvic pain, neuroma pain, complex regional pain syndrome, pain associated with gastrointestinal distension, chronic arthritic pain and related neuralgias, and pain associated with cancer, pain associated with chemotherapy, HIV and HIV treatment-induced neuropathy; and pain associated with diseases or disorders selected from the group consisting of hyperalgesia, allodynia, irritable bowel syndrome..

20. Use according to claims 12, 15 or 16, wherein said disease is dysmenorrhea,

dyspareunia, endometriosis, adenomyosis, endometriosis-associated pain, or other endometriosis-associated symptoms, wherein said symptoms are in particular endometriosis-associated proliferation, dysmenorrhea, dyspareunia, dysuria, or dyschezia. Use according to claims 12. 15 or 16. for the treatment of inflammatory bowel disease (ulcerative colitis and Crohn's disease), hyperaemia, sepsis, metabolic disorders, e.g. obesity, insulin resistance, diabetes mellitus type 1 and 2, metabolic endocrine disorder; metabolic syndrome; atherosclerosis, reperfusion injury, inflammatory bone resorption, inflammatory liver diseases, pulmonary fibrosis, acute respiratory distress syndrome, and intestinal polyposis, inflammatory skin diseases like psoriasis, pemphigus vulgaris, inflammatory eye disorders like non-infectious uveitis, primary and secondary autoimmune uveitis, VKH syndrome, anterior uveitis, intermediate uveitis, posterior uveitis, panuveitis, Behcet's disease and neuromyelitis optica.fibrotic diseases like idiopathic pulmonary fibrosis, skin fibrosis, systemic sclerosis, autism disorders, liver diseases like nonalcoholic-, alcoholic- and toxic fatty liver disease, steatohepatitis, hepatic fibrosis; and cirrhosis; lung diseases like chronic obstructive pulmonary disease, asthma, pneumonia; neurodegenerative diseases like Parkinson's disease, Alzheimer's disease; stroke, postischemic brain injury, brain ischemia, posttraumatic brain injury, alopecia, acute coronary syndrome, myocardial infarction, autoimmune diseases like autoimmune encephalomyelitis, multiple sclerosis; arthritis (such as osteoarthritis and rheumatoid arthritis); , psoriatic arthritis, ankylosing spondylitis), psoriasis, lupus erythematosus (e.g. systemic lupus erythematosus, cutaneous lupus and neonatal lupus erythematosus); inflammatory, autoimmune and fibrotic kidney diseases (e.g. glomerulonephritis, lupus nephritis, ANCA) interstitial cystitis, hypertrophy of the e.g. kidney, ischemia/reperfusion injury, allergic rhinitis, burn wound, osteoporosis viral and bacterial infections, chemotherapy-induced alopecia, cachexia induced for any reason, e.g. cancer, heart failure, etc.. muscular atrophy, pancreatitis, schizophrenia, seizures, epilepsy, Fragile X syndrome, graft- versus-host disease, graft rejection, heart fibrosis, autoimmune myocardial disease, myocarditis, endocarditis, ischemia-reperfusion injury following e.g. myocardial infarction, hypertension, artherosclerosis, acute lung injury, ARDS, hypersensitivity pneumonitis, lung fibrosis, e.g. idiopathic pulmonary fibrosis, lymphocytic bronchiolitis e.g. after lung transplantation, dry eye, subfertility (e.g. associated with inflammatory conditions such as endometriosiso, or metabolic endocrine disorders)

Description:
SUBSTITUTED PYRAZOLOPYRIDINAMINES

The present invention relates to substituted pyrazolopyridinamine compounds of general formula (I) as described and defined herein, to methods of preparing said compounds, to intermediate compounds useful for preparing said compounds, to pharmaceutical compositions and combinations comprising said compounds and to the use of said compounds for manufacturing a pharmaceutical composition for the treatment or prophylaxis of a disease, in particular of a hyperproliferative, angiogenesis disorders, inflammatory diseases or diseases associated with inflammatory pain, as a sole agent or in combination with other active ingredients.

BACKGROUND OF THE INVENTION

The present invention relates to chemical compounds that inhibit MKNK1 kinase (also known as MAP Kinase interacting Kinase, Mnk1 ) and/or MKNK2 kinase (also known as MAP Kinase interacting Kinase, Mnk2). Human MKNKs comprise a group of four proteins encoded by two genes (Gene symbols: MKNK1 and MKNK2) by alternative splicing. The b-forms lack a MAP kinase-binding domain situated at the C-terminus. The catalytic domains of the MKNK1 and MKNK2 are very similar and contain a unique DFD (Asp-Phe-Asp) motif in subdomain VII, which usually is DFG (Asp-Phe-Gly) in other protein kinases and suggested to alter ATP binding [Jauch et al., Structure 13, 1559-1568, 2005 and Jauch et al., EMBO J25,

4020-4032, 2006]. MKNKI a binds to and is activated by ERK and p38 MAP Kinases, but not by JNK1 . MKNK2a binds to and is activated only by ERK. MKNK1 b has low activity under all conditions and MKNK2b has a basal activity independent of ERK or p38 MAP Kinase.

[Buxade M et al., Frontiers in Bioscience 5359-5374, May 1 , 2008]

MKNKs have been shown to phosphorylate eukaryotic initiation factor 4E (elF4E), heterogeneous nuclear RNA-binding protein A1 (hnRNP A1 ), polypyrimidine-tract binding protein-associated splicing factor (PSF), cytoplasmic phospholipase A2 (cPLA2) and Sprouty 2 (hSPRY2) [Buxade M et al., Frontiers in Bioscience 5359-5374, May 1 , 2008].

elF4E is an oncogene that is amplified in many cancers and is phosphorylated exclusively by MKNKs proteins as shown by KO-mouse studies [Konicek et al., Cell Cycle 7: 16, 2466-2471 , 2008; Ueda et al., Mol Cell Biol 24, 6539-6549, 2004]. elF4E has a pivotal role in enabling the translation of cellular mRNAs. elF4E binds the 7-methylguanosine cap at the 5 ' end of cellular mRNAs and delivers them to the ribosome as part of the elF4F complex, also containing elF4G and elF4A. Though all capped mRNAs require elF4E for translation, a pool of mRNAs is exceptionally dependent on elevated elF4E activity for translation. These so-called "weak mRNAs" are usually less efficiently translated due to their long and complex 5 ' UTR region and they encode proteins that play significant roles in all aspects of malignancy including VEGF, FGF-2, c-Myc, cyclin D1 , survivin, BCL-2, MCL-1 , MMP-9, heparanase, etc. Expression and function of elF4E is elevated in multiple human cancers and directly related to disease progression [Konicek et al., Cell Cycle 7: 16, 2466-2471 , 2008].

MKNK1 and MKNK2 are the only kinases known to phosphorylate elF4E at Ser209. Overall translation rates are not affected by elF4E phosphorylation, but it has been suggested that elF4E phosphorylation contributes to polysome formation (i.e. multiple ribosome on a single mRNA) that ultimately enables more efficient translation of "weak mRNAs" [Buxade M et al., Frontiers in Bioscience 5359-5374, May 1 , 2008]. Alternatively, phosphorylation of elF4E by MKNK proteins might facilitate elF4E release from the 5' cap so that the 48S complex can move along the "weak mRNA" in order to locate the start codon [Blagden SP and Willis AE, Nat Rev Clin Oncol. 8(5):280-91 , 201 1 ]. Accordingly, increased elF4E phosphorylation predicts poor prognosis in non-small cell lung cancer patients [Yoshizawa et al., Clin Cancer Res. 16(1 ):240-8, 2010]. Further data point to a functional role of MKNK 1 in carcinogenesis, as overexpression of constitutively active MKNK1 , but not of kinase-dead MKNK1 , in mouse embryo fibroblasts accelerates tumor formation [Chrestensen C. A. et al., Genes Cells 12, 1 133-1 140, 2007]. Moreover, increased phosphorylation and activity of MKNK proteins correlate with overexpression of HER2 in breast cancer [Chrestensen, C. A. et al., J. Biol. Chem. 282, 4243-4252, 2007]. Constitutively active, but not kinase-dead, MKNK1 also accelerated tumor growth in a model using Εμ-Myc transgenic hematopoietic stem cells to produce tumors in mice. Comparable results were achieved when an elF4E carrying a S209D mutation was analyzed. The S209D mutation mimicks a phosphorylation at the MKNK1 phosphorylation site. In contrast, a non-phosphorylatable form of elF4E attenuated tumor growth [Wendel HG, et al., Genes Dev. 21 (24):3232-7, 2007]. A selective MKNK inhibitor that blocks elF4E phosphorylation induces apoptosis and suppresses proliferation and soft agar growth of cancer cells in vitro. This inhibitor also suppresses outgrowth of experimental B16 melanoma pulmonary metastases and growth of subcutaneous HCT1 16 colon carcinoma xenograft tumors without affecting body weight [Konicek et al., Cancer Res. 71 (5): 1849-57, 201 1 ]. In summary, elF4E phosphorylation through MKNK protein activity can promote cellular proliferation and survival and is critical for malignant transformation.

Inhibition of MKNK activity may provide a tractable cancer therapeutic approach.

Furthermore it has been found that MKNK1 is an acinar cell-specific kinase required for exocrine pancreatic secretion [Cendrowski J, Sanchez-Arevalo Lobo VJ, Sendler M, et al. Gut Published Online First: July 18, 2014; doi: 10.1 136/gutjnl-2013-306068]. The kinases KNK1 and MKNK2 are important downstream targets of the Erk and p38 mitogen-activated protein kinase (MARK) pathways and their activity can also be modulated by MARK independent signals. The MKNKs are directly involved in regulating mRNA translation and, therefore, are key mediators of oncogenic progression and cytokine signaling. In particular, MARK pathways such as Erk and p38 have been shown to play important roles in modulating immune responses by mediating the production of cytokines that control the initiation of innate immunity; the activation of adaptive immunity; and by regulating cellular responses to cytokines involved in immune responses. In addition, Erk and p38 contribute to pain sensitivity and p38 kinase inhibitors have shown pre-clinical and clinical efficacy regarding pain [Brown, Heitmeyer, et al., J Inflamm (Lond), 2008; Hill,

Dabbagh, et al., J Pharmacol Exp TherJi, 2008; Gereau, et al., Brain Res Rev, 2009; Cheng, Dauch, et al., ol Pain, 2010; Anand, Shenoy, et al., European Journal of Pain, 201 1 ;

Daves, Aitchison, et al., American College of Rheumatology Annual Meeting, 2012; Lin, Wang, et al., Curr Med Chem, 2014]. As MKNK kinases are effectors of MAPK pathways, these observations suggest that they may play important roles in mediating cytokine production and inflammatory pain. Recent studies support the involvement of MKNK kinases in different inflammatory processes [Rowlett, Chrestensen, et al., Am J Physiol Gastrointest Liver Physiol, 2008; Kjellerup, Kragballe, et al., Experimental Dermatology, 2008;

Melemedjian, Asiedu, et al., J Neurosci, 2010; Fortin, Mayer, et al., Journal of Leukocyte Biolog, 2013]. Due to the induction of MKNK kinases by different inflammatory stimuli (sterile inflammation and pathogens) and their ability to regulate the expression of different cytokines which mediate the pathogenesis of multiple disorders such as auto-immune diseases, allergies, neurological disorders, sepsis, cardiovascular diseases, metabolic diseases, obesity and cancer. MKNKs represent a central node in regulating inflammation. [Joshi et al.; World J Biol Chem 2014 August 26; 5(3): 321-333; Joschi et al., Biomol Concepts. 2012 April ; 3(2): 127-139]

Imbalance in cytokines from lnterleukin-1 family and their role in the pathogenesis of Endometriosis has been reported in the literature [American Journal of Reproductive

Immunology 68 (2012) 138-145] as well as the possible pathophysiological roles of Mitogen- Activated Protein Kinases (MAPKs) in Endometriosis [Yoshino et al.; AJRI 2004: 52: 306- 311]. More recently, the role of pro-inflammatory cytokines for evaluation of inflammatory status and their pathogenetic mechanisms in endometriosis has been illustrated [Tosti et al.; Reproductive Sciences 2015, 1-7; Malutan et al., Centr Eur J Immunol 2015: 40 (1): 96-102: Soo Hyun Ahn et al., BioMed Research International, Vol. 2015, Article ID 795976, 12 pages]. Women with endometriosis have elevated levels of key pro-inflammatory cytokines, i.e. IL-1 β, IL-6, and TNF-a. At the same time, IL-1 β and IL-6 could be used as predictors for endometriosis.

Substituted pyrazolopyridinamine compounds of general formula (I) have not been disclosed in prior art for the treatment or prophylaxis of different diseases.

So, the state of the art described above does not describe the specific substituted pyrazolopyridinamine compounds of general formula (I) of the present invention as defined herein or a stereoisomer, a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same, as described and defined herein, and as hereinafter referred to as

"compounds of the present invention", or their pharmacological activity.

It has now been found, and this constitutes the basis of the present invention, that said compounds of the present invention have surprising and advantageous properties.

In particular, said compounds of the present invention have been found to effectively inhibit KNK1 kinase.

Furthermore, the compounds according to the present invention have been found to effectively inhibit KNK2 kinase.

In contrast to other MKNK1 and/or MKNK2 kinase inhibitors, the pyrazolopyridinamines according to the invention are mainly active on sterile and pathogenic inflammatory responses and do not interfere directly with cell viability.

The pyrazolopyridinamines according to the present invention may be used for the treatment or prophylaxis of diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses or diseases which are accompanied with uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, particularly in which the uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses is mediated by MKNK1 and/or KNK2 kinase, such as, for example, haematological tumours, solid tumours, and/or metastases thereof, e.g. leukaemias and myelodysplastic syndrome, malignant lymphomas, head and neck tumours including brain tumours and brain metastases, tumours of the thorax including non-small cell and small cell lung tumours, gastrointestinal tumours, endocrine tumours, mammary and other gynaecological tumours, urological tumours including renal, bladder and prostate tumours, skin tumours, and sarcomas, and/or metastases thereof.

The pyrazoiopyridinamines according to the present invention may be used for the treatment or prophylaxis of inflammatory and/or immunological diseases as described in the summary of the invention.

Furthermore, the compounds according to the invention may be used for the treatment or prophylaxis of a gynecological disease, preferably dysmenorrhea, dyspareunia or endometriosis, adenomyosis, endometriosis-associated pain, or other endometriosis- associated symptoms, wherein said symptoms are in particular endometriosis-associated proliferation, dysmenorrhea, dyspareunia, dysuria, or dyschezia.

SUMMARY OF THE INVENTION

The present invention covers com ounds of general formula (I) :

in which :

R 1 represents a halogen atom or a group selected from:

hydroxy-, cyano-, d-Ce-alkyl-, Ci-Ce-alkoxy-, halo-Ci-Ce-alkyk halo-Ci-Ce-alkoxy-, hydroxy-Ci-Ce-alkyl-, C i -C 3 -a I koxy-C i -C 3 -a I ky I- , S-(Ci-Cr-alkyl)

R 2 represents a hydrogen atom or a group d-Ce-alkyl-;

R 3 represents a group selected from: -C(=0)-0-R 5 , -C(=0)-N(R 5a )R 5 ;

R 4 represents halo-, hydroxy-, oxo- (0=), cyano-, nitro-, Ci-Ce-alkyl-, C2-Ce-alkenyl-,

C2-Ce-alkynyl-, halo-C--Ce-alkyl-, Ci-Ce-alkoxy-, halo-Ci-Ce-alkoxy-,

hyd roxy-C -Ce-a I ky I-, Ci-Ce-alkoxy-Ci-Ce-alkyl-, halo-Ci-Ce-alkoxy-C -Ce-alkyl-,

R 6 -0-, -C(=0)-R 6 , -C(=0)-0-R 6 , -0-C(=0)-R 6 , -N(R 6a )-C(=0)-R 6b , -N(R 6a )-C(=0)-0-

R 6b , -N(R 6a )-C(=0)-N(R 6b )R 6c , -N(R 6a )R 6b , -N(R 6a )R 6d , -C(=0)-N(R 6a )R 6b , R 6 -S-,

R 6 -S(=0)-, R 6 -S(=0) 2 -, -N(R 6a )-S(= D6b °(=0)-N(R 6a )R 6b , -N(R 6a )-S(=0) 2 - R 6b , -S(=0) 2 -N(R 6a )R 6b , -S(=0)=N(R 6a )R 6b , -N=S(=0)(R 6a )R 6b or -(Ci-Ce-alkyl)-

N(R 6a )R 6b ;

R 5 , R 5a , R 5b are the same or different and independently selected from each other, and represent a hydrogen atom or a group selected from: Ci-Ce-alkyl-, Cs-Ce-cycloalkyk

3- to 10-membered heterocycloalkyi-, aryl-, and heteroaryl-;

wherein said Ci-Ce-alkyl-, Cs-Ce-cycloalkyl-, 3- to 10-membered heterocycloalkyi-, aryl- and heteroaryl- group is optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups ;or

N(R 5a )R 5 together represent a 3- to 10-membered heterocycloalkyi- group, said group being optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups ;

R 6 , R 6a , R 6b , R 6c are the same or different and independently selected from each other, and represent a hydrogen atom, a Ci-Ce-alkyl- or a Cs-Ce-cycloalkyl- group; or

R 6a and R 6b , or

R 6a and R 6c , or

R 6b and R 6c together may form a C2-Ce-alkylene group, in which optionally one methylene can be replaced by -0-, -C(=0)-, -NH-, or -N(CrC 4 -alkyl)- ;

R 6d represents -(Ci-C 6 -alkyl)-N(R 6a )R 6b ; or a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.

The present invention further relates to methods of preparing compounds of general formula (I), to pharmaceutical compositions and combinations comprising said compounds, to the use of said compounds for manufacturing a pharmaceutical composition for the treatment or prophylaxis of a disease, as well as to intermediate compounds useful in the preparation of said compounds. DETAILED DESCRIPTION OF THE INVENTION

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, preferably a fluorine, chlorine or bromine atom.

The term "Ci-Cio-alkyl" is to be understood as preferably meaning a linear or branched, saturated, monovalent hydrocarbon group havinn 1 2, 3, 4, 5, 6, 7, 8, 9 or 10 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, 4, 5 or 6 carbon atoms ("CrCe-alkyl"), more particularly, said group has 1 , 2, 3 or 4 carbon atoms ("Ci-C4-alkyl"), e.g. a methyl, ethyl, propyl, butyl, iso-propyl, iso-butyl, sec-butyl, tert-butyl group; even more particularly 1 . 2 or 3 carbon atoms ("Ci-Cs-alkyr), e.g. a methyl, ethyl, n-propyl- or iso-propyl group.

The term "Ci-Cio-alkylene" is to be understood as preferably meaning a linear or branched, saturated, bivalent hydrocarbon group having 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms, e.g. a methylene, ethylene, n-propylene, n-butylene, n-pentylene, 2-methylbutylene, n-hexylene,

3- methylpentylene group, or an isomer thereof. Particularly, said group is linear and has 2, 3, 4 or 5 carbon atoms ("Ca-Cs-alkyiene"), e.g. an ethylene, n-propylene, n-butylene, n- pentylene group, more particularly 3 or 4 carbon atoms ("Cs-Ci-alkylene"), e.g. an n- propylene or n-butylene group.

The term "halo-d-Ce-alkyl" is to be understood as preferably meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "d-Ce-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 halo-Ci-Ce-alkyl group is, for example, -CF:,, -CHF 2 , -CH2F, -CF2CF3 or

The term "Ci-Ce-alkoxy" is to be understood as preferably meaning a linear or branched, saturated, monovalent, hydrocarbon group of formula -O-(Ci-Ce-alkyl), in which the term "Ci- Ce-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.

The term "halo-Ci-Ce-alkoxy" is to be understood as preferably meaning a linear or branched, saturated, monovalent Ci-Ce-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 halo-Ci-Ce-alkoxy group is, for example,

-OCF3, -OCHF2, -OCH2F, -OCF2CF3 or -OCH2CF3. The term "C i -Ce-a I koxy-C i -Ce-a I ky Γ is to be understood as preferably meaning a linear or branched, saturated, monovalent Ci-Ce-alkyl group, as defined supra, in which one or more of the hydrogen atoms is replaced, in identically or differently, by a d-Ce-alkoxy group, as defined supra, e.g. methoxyalkyl, ethoxyalkyl, propyloxyalkyl, iso-propoxyalkyl, butoxyalkyl, iso-butoxyalkyl, tert-butoxyalkyl, sec-butoxyalkyl, pentyloxyalkyl, iso-pentyloxyalkyl, hexyloxyalkyl group, or an isomer thereof.

The term "halo-Ci-Ce-alkoxy-Ci-Ce-alkyr is to be understood as preferably meaning a linear or branched, saturated, monovalent C i -Ce-a I koxy-C i -Ce-a I ky I 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 h a lo-C -Ce-a I koxy-C i -Ce-a I ky I group is, for example, -CH 2 CH 2 OCF 3 , -CH 2 CH 2 OCHF 2 , -CH 2 CH 2 OCH 2 F, -CH 2 CH 2 OCF 2 CF 3 or The term "C 2 -Cio-alkenyl" is to be understood as preferably meaning a linear or branched, monovalent hydrocarbon group, which contains one or more double bonds, and which has 2, 3, 4, 5, 6, 7. 8, 9 or 10 carbon atoms, particularly 2, 3, 4, 5 or 6 carbon atoms

("C 2 -Ce-alkenyl"), more particularly 2 or 3 carbon atoms ("Cz-Cs-alkenyl"), it being understood that in the case in which said alkenyl group contains more than one double bond, then said double bonds may be isolated from, or conjugated with, each other. Said alkenyl group is, for example, a vinyl, allyl, (£)-2-methylvinyl, (Z)-2-methylvinyl, homoallyl, (E)-but-2-enyl, (Z)-but-2-enyl, (E)-but-1 -enyl, (Z)-buM -enyl, pent-4-enyl, (E)-pent-3-enyl, (Z)-pent-3-enyl, (E)-pent-2-enyl, (Z)-pent-2-enyl, (E)-pent-l -enyl, (Z)-pent-l -enyl, hex-5-enyl, (E)-hex-4-enyl, (Z)-hex-4-enyl, (E)-hex-3-enyl, (Z)-hex-3-enyl, (E)-hex-2-enyl, (Z)-hex-2-enyl, (E)-hex-l -enyl, (Z)-hex-l -enyl, /so-propenyl, 2-methylprop-2-enyl, 1 -methylprop-2-enyl, 2-methylprop-1 -enyl, (E)-1 -methylprop-1 -enyl, (Z)-1 -methylprop-1 -enyl, 3-methylbut-3-enyl, 2-methylbut-3-enyl, 1 -methylbut-3-enyl, 3-methylbut-2-enyl, (E)-2-methylbut-2-enyl, (Z)-2-methylbut-2-enyl, (E)-1 -methylbut-2-enyl, (Z)-1 -methylbut-2-enyl, (E)-3-methylbut-1 -enyl,

(Z)-3-methylbut-1 -enyl, (E)-2-methylbut-1 -enyl, (Z)-2-methylbut-1 -enyl,

(E)-1 -methylbut-1 -enyl, (Z)-1 -methylbut-1 -enyl, 1 .1 -dimethylprop-2-enyl, 1 -ethylprop-1 -enyl,

1 - propylvinyl, 1 -isopropylvinyl, 4-methylpent-4-enyl, 3-methylpent-4-enyl,

2- methylpent-4-enyl, 1 -methylpent-4-enyl, 4-methylpent-3-enyl, (E)-3-methylpent-3-enyl, (Z)-3-methylpent-3-enyl, (E)-2-methylpent-3-enyl, (Z)-2-methylpent-3-enyl,

(E)-1 -methylpent-3-enyl, (Z)-1 -methylpent-3-enyl, (E)-4-methylpent-2-enyl,

(Z)-4-methylpent-2-enyl, (E)-3-methylpent-2-enyl, (Z)-3-methylpent-2-enyl,

(E)-2-methylpent-2-enyl, (Z)-2-methylpent-2-enyl, (E)-1 -methylpent-2-enyl,

(Z)-1 -methylpent-2-enyl, (E)-4-methylpent-1 -enyl, (Z)-4-methylpent-1 -enyl, (E)-3-methylpent-1 -enyl. (Z)-3-methylpent-1 -enyl, (E)-2-meihylpeni-1 -enyl, (Z)-2-methylpent-1 -enyl, (E)-1 -methylpent-1 -enyl, (Z)-1 -methylpent-1 -enyl, 3-ethylbut-3-enyl, 2-ethylbut-3-enyl, 1 -ethylbut-3-enyl, (E)-3-ethylbut-2-enyl, (Z)-3-ethylbut-2-enyl,

(E)-2-ethylbut-2-enyl, (Z)-2-eihylbut-2-enyl, (E)-1 -ethylbut-2-enyl, (Z)-1 -ethylbut-2-enyl, (E)-3-ethylbut-1 -enyl, (Z)-3-ethylbut-1 -enyl, 2-ethylbut-1 -enyl, (E)-1 -ethylbui-1 -enyl,

(Z)- 1 -ethylbut- 1 -enyl , 2-propylprop-2-enyl, 1 -propylprop-2-enyl, 2-isopropylprop-2-enyl, 1 -isopropylprop-2-enyl, (E)-2-propyiprop-1 -enyl, (Z)-2-propylprop-1 -enyl,

(E)-1 -propylprop-1 -enyl, (Z)-1 -propylprop-1 -enyl, (E)-2-isopropylprop-1 -enyl,

(Z)-2-isopropylprop-1 -enyl, (E)-1 -isopropylprop-1 -enyl, (Z)-1 -isopropylprop-1 -enyl,

(E)-3,3-dimethylprop-1 -enyl, (Z)-3,3-dimeihylprop-1 -enyl, 1 -(1 , 1 -dimethylethyl)ethenyl, buta-1 ,3-dienyl, penta-1 ,4-dienyl, hexa-1 ,5-dienyl, or methylhexadienyl group. Particularly, said group is vinyl or allyl.

The term "C2-Cio-alkynyl" is to be understood as preferably meaning a linear or branched, monovalent hydrocarbon group which contains one or more triple bonds, and which contains 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms, particularly 2, 3, 4, 5 or 6 carbon atoms

("C2-Ce-alkynyl"), more particularly 2 or 3 carbon atoms ("C2-C3-alkynyl"). Said C2-Cio-alkynyl group is, for example, ethynyl, prop-1 -ynyl, prop-2-ynyl, but-1 -ynyl, but-2-ynyl, but-3-ynyl, pent-1 -ynyl, pent-2-ynyl, pent-3-ynyl, pent-4-ynyl, hex-1 -ynyl, hex-2-ynyl, hex-3-ynyl, hex-4-ynyl, hex-5-ynyl, 1 -methylprop-2-ynyl, 2-methylbut-3-ynyl, 1 -methylbut-3-ynyl,

1 - methylbut-2-ynyl, 3-methylbut-1 -ynyl, 1 -ethylprop-2-ynyl, 3-methylpent-4-ynyl,

2- methylpent-4-ynyl, 1 -methylpent-4-ynyl, 2-methylpent-3-ynyl, 1 -methylpent-3-ynyl, 4-methylpent-2-ynyl, 1 -methylpent-2-ynyl, 4-methylpent-1 -ynyl, 3-methylpent-1 -ynyl, 2-ethylbut-3-ynyl, 1 -ethylbut-3-ynyl, 1 -ethylbut-2-ynyl, 1 -propylprop-2-ynyl,

1 -isopropylprop-2-ynyl, 2,2-dimethylbut-3-ynyl, 1 , 1-dimethylbut-3-ynyl,

1 , 1 -dimethylbut-2-ynyl, or 3,3-dimethylbut-1 -ynyl group. Particularly, said alkynyl group is ethynyl, prop-1 -ynyl, or prop-2-ynyl.

The term "Cs-Cio-cycloalkyl" is to be understood as meaning a saturated, monovalent, mono-, or bicyclic hydrocarbon ring which contains 3, 4, 5, 6, 7, 8. 9 or 10 carbon atoms

("Cs-Cio-cycloalkyl"). Said Cs-Cio-cycloalkyl group is for example, a monocyclic hydrocarbon ring, e.g. a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl or cyclodecyl, or a bicyclic hydrocarbon ring, e.g. a perhydropentalenylene or decalin ring. Particularly, said ring contains 3, 4, 5 or 6 carbon atoms CCs-Ce-cycloalkyl"). The term "Cs-Ce-cycloalkyloxy " refers to a (Cn-Ce-cycloa I kyl )-0- group in which

"Cs-Ce-cycloalkyr is as defined herein. Examples include, but are not limited to,

cyclopropanoxy and cyclobutanoxy. The term "C4-Cic-cycloalkenyl " is to be understood as preferably meaning a non-aromatic, monovalent, mono- or bicyclic hydrocarbon ring which contains 4, 5, 6, 7, 8, 9 or 10 carbon atoms and one, two, three or four double bonds, in conjugation or not, as the size of said cycloalkenyl ring allows. Said C-j-Cic-cycloalkenyl group is for example, a monocyclic hydrocarbon ring, e.g. a cyclobutenyl, cyclopentenyl, or cyclohexenyl or a bicyclic hydrocarbon, e.g. :

The term "Cs-Cs-cycloalkenyloxy" refers to a (Cs-Ce-cycloa I ke nyl )-0- group in which

"Cs-Ce-cycloalkenyl" is as defined herein.

The term "3- to 10-membered heterocycioalkyi", is to be understood as meaning a saturated, monovalent, mono- or bicyclic hydrocarbon ring which contains 2, 3, 4, 5, 6, 7, 8 or 9 carbon atoms, and one or more heteroatom-containing groups selected from -C(=0)-, -0-, -S-, -S(=0)-, -S(=0)2-, -N( a )-, in which R a represents a hydrogen atom or a Ci-Ce-alkyl group; it being possible for said heterocycioalkyi group to be attached to the rest of the molecule via any one of the carbon atoms or, if present, the nitrogen atom. Heterospirocycloalkyl, heterobicycioalkyi and bridged heterocycioalkyi, as defined infra, are also included within the scope of this definition.

The term "heterospirocycloalkyl" is to be understood as meaning a saturated, monovalent bicyclic hydrocarbon radical in which the two rings share one common ring carbon atom, and wherein said bicyclic hydrocarbon radical contains 2, 3, 4, 5, 6, 7, 8 or 9 carbon atoms, and one or more heteroatom-containing groups selected from C(=0), O, S, S(=0), NR a , in which R a represents a hydrogen atom or a Ci-Ce-alkyl- or Cs-Cz-cycloalkyl- group; it being possible for said heterospirocycloalkyl- group to be attached to the rest of the molecule via any one of the carbon atoms or, if present, the nitrogen atom. Said heterospirocycloalkyl- group is, for example, azaspiro[2.3]hexyl-, azaspiro[3.3]heptyl-, oxaazaspiro[3.3]heptyl-, thiaazaspiro[3.3]heptyl-, oxaspiro[3.3]heptyl-, oxazaspiro[5.3]nonyl-, oxazaspiro[4.3]octyl-, oxazaspiro[5.5]undecyl-, diazaspiro[3.3]heptyl-, thiazaspiro[3.3]heptyl-, thiazaspiro[4.3]octyl-, or azaspiro[5.5]decyl-.

The term "heterobicycloalkyl" is to be understood as meaning a saturated, monovalent bicyclic hydrocarbon radical in which the two rings share two immediately adjacent ring atoms, and wherein said bicyclic hydrocarbon radical contains 2, 3, 4, 5, 6, 7, 8 or 9 carbon atoms, and one or more heteroatom-containing groups selected from C(=0), O, S, S(=0), S(=0)2, NR a , in which R a represents a hydrogen atom or a Ci-Ce-alkyl- or Cs-Cr-cycloalkyl- group: it being possible for said heterobicycloalkyl- group to be attached to the rest of the molecule via any one of the carbon atoms or, if present, the nitrogen atom. Said

heterobicycoalkyl- group is, for example, azabicyclo[3.3.0]octyl-, azabicyclo[4.3.0]nonyl-, diazabicyclo[4.3.0]nonyl-, oxazabicyclo[4.3.0]nonyl-, thiazabicyclo[4.3.0]nonyl-, or azabicyclo[4.4.0]decyl-. The term "bridged heterocycioalkyi" is to be understood as meaning a saturated, monovalent bicyclic hydrocarbon radical in which the two rings share two common ring atoms which are not immediately adjacent, and wherein said bicyclic hydrocarbon radical contains 2, 3, 4, 5, 6, 7, 8 or 9 carbon atoms, and one or more heteroatom-containing groups selected from C(=0), O, S, S(=0), S(=0) 2 , NR a , in which R a represents a hydrogen atom, or a

Ci-Ce-alkyl- or Cs-Cz-cycloalkyl- group: it being possible for said bridged heterocycloalkyl- group to be attached to the rest of the molecule via any one of the carbon atoms or, if present, the nitrogen atom. Said bridged heterocycioalkyi- group is, for example, azabicyclo[2.2.1 jheptyl-, oxazabicyclo[2.2.1 jheptyl-, thiazabicyclo[2.2.1 jheptyl-,

diazabicyclo[2.2.1 jheptyl-, azabicyclo[2.2.2]octyl-, diazabicyclo[2.2.2]octyl-,

oxazabicyclo[2.2.2]octyl-, thiazabicyclo[2.2.2]octyl-, azabicyclo[3.2.1 joctyl-,

diazabicyclo[3.2.1 joctyl-, oxazabicyclo[3.2.1 joctyl-, thiazabicyclo[3.2.1 joctyl-,

azabicyclo[3.3.1 jnonyl-, diazabicyclo[3.3.1 jnonyl-, oxazabicyclo[3.3.1 jnonyl-,

thiazabicyclo[3.3.1 jnonyl-, azabicyclo[4.2.1 jnonyl-, diazabicyclo[4.2.1 jnonyl-,

oxazabicyclo[4.2.1 jnonyl, thiazabicyclo[4.2.1 jnonyl-, azabicyclo[3.3.2]decyl-,

diazabicyclo[3.3.2]decyk oxazabicyclo[3.3.2jdecyl-, thiazabicyclo[3.3.2]decyl-, or azabicyclo[4.2.2]decyl-.

Particularly, said 3- to 10-membered heterocycioalkyi can contain 2, 3, 4, or 5 carbon atoms, and one or more of the above-mentioned heteroatom-containing groups (a "3- to

6-membered heterocycioalkyi"), more particularly said 3- to 10-membered heterocycioalkyi can contain 4 or 5 carbon atoms, and one or more of the above-mentioned

heteroatom-containing groups (a "5- to 6-membered heterocycioalkyi"). Particularly, without being limited thereto, said 3- to 10-membered heterocycloalkyi can be a 4-membered ring, such as an azetidinyl, oxetanyl, or a 5-membered ring, such as tetrahydrofuranyl, dioxolinyl, pyrrol id inyl, imidazolidinyl, pyrazolidinyl or a 6-membered ring, such as tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, or trithianyl, or a 7-membered ring, such as a diazepanyl ring, for example.

Said 3- to 10-membered heterocycloalkyi can be bicyclic, such as, without being limited thereto, a 5, 5-membered ring, e.g. a hexa hyd rocyclopenta [c] pyrrol-2( 1 H )-yl ring, or a 5, 6-membered bicyclic ring, e.g. a hexahydropyrrolo[1 ,2-a]pyrazin-2(1 H)-yl ring.

The term "4- to 10-membered heterocycloalkenyl", is to be understood as meaning an non- aromatic, unsaturated, monovalent, mono- or bicyclic hydrocarbon ring which contains 3, 4, 5, 6, 7, 8 or 9 carbon atoms, and one or more heteroatom-containing groups selected from -C(=0)-, -0-, -S-, -S(=0)-, -S(=0) 2 -, -N(R a )-, in which R a represents a hydrogen atom or a Ci-Ce-alkyl group; it being possible for said heterocycloalkenyl group to be attached to the rest of the molecule via any one of the carbon atoms or, if present, the nitrogen atom.

Examples of said heterocycloalkenyl are e.g. 4H-pyranyl, 2H-pyranyl. 3H-diazirinyl,

2,5-dihydro-1 H-pyrrolyl, [1 ,3]dioxolyl, 4H-[1 .3,4]thiadiazinyl, 2,5-dihydrofuranyl,

2,3-dihydrofuranyl, 2,5-dihydrothiophenyl, 2,3-dihydrothiophenyl, 4,5-dihydrooxazolyl, or 4H-[1 ,4]thiazinyl group.

The term "aryS" is to be understood as preferably meaning a monovalent, aromatic or partially aromatic, mono-, bi- or tricyclic hydrocarbon ring having 6, 7, 8, 9, 10, 1 1 . 12, 13 or 14 carbon atoms (a "C6-Ci4-aryl" group), particularly a ring having 6 carbon atoms (a "Ce-aryl" group), e.g. a phenyl group; or a biphenyl group, or a ring having 9 carbon atoms (a "Cg-aryl" group), e.g. an indanyl or indenyl group, or a ring having 10 carbon atoms (a "Cio-aryl" group), e.g. a tetralinyl, dihydronaphthyl, or naphthyl group, or a ring having 13 carbon atoms, (a "Ci3-aryl" group), e.g. a fluorenyl group, or a ring having 14 carbon atoms, (a

"Ci4-aryl" group), e.g. an anthranyl group. Preferably, the aryl group is a phenyl group.

The term "heteroaryl" is understood as preferably meaning a monovalent, monocyclic, bicyclic or tricyclic aromatic ring system having 5, 6, 7, 8, 9, 10, 1 1 , 12, 13 or 14 ring atoms (a "5- to 14-membered heteroaryl" group), particularly 5 or 6 or 9 or 10 atoms, and which contains at least one heteroatom which may be identical or different, said heteroatom being such as oxygen, nitrogen or sulfur, and in addition in each case can be benzocondensed. Particularly, heteroaryl is selected from thienyl, furanyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl,

thia-4H-pyrazolyl efc, and benzo derivatives thereof, such as, for example, benzofuranyl, benzothienyl, benzoxazolyl, benzisoxazolyl, benzimidazolyl, benzotriazolyl, indazolyl, indolyl, isoindolyl, efc; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, efc, and benzo derivatives thereof, such as, for example, quinolinyl, quinazolinyl, isoquinolinyl. efc; or azocinyl, indolizinyl, purinyl, efc, and benzo derivatives thereof; or cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthpyridinyl, pteridinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, xanthenyl, or oxepinyl, efc. In general, and unless otherwise mentioned, the heteroarylic or heteroarylenic radicals include all the possible isomeric forms thereof, e.g. the positional isomers thereof. Thus, for some illustrative non-restricting example, the term pyridinyl or pyridinylene includes pyridin-2-yl, pyridin-2-ylene, pyridin-3-yl, pyridin-3-ylene, pyridin-4-yl and pyridin-4-ylene; or the term thienyl or thienylene includes thien-2-yl, thien-2-ylene, thien-3-yl and thien-3-ylene.

The term "d-d", as used throughout this text, e.g. in the context of the definition of "d-d-alkyl", "d-d-haloalkyl", "d-d-alkoxy", or "d-d-haloalkoxy" is to be understood as meaning an a Iky I 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 "d-d" is to be interpreted as any sub-range comprised therein, e.g. d- , d-d , d- , d-d , d-d , d- , d-d ; particularly d-d , d-d , d-d , d-d. d-d ; more particularly d-d ; in the case of "d-d-haloalkyl" or "d-d-haloalkoxy" even more particularly d-d.

Similarly, as used herein, the term "d-d", as used throughout this text, e.g. in the context of the definitions of "d-d-alkenyl" and "d-d-alkynyl". is to be understood as meaning an alkenyl group or an alkynyl 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 "d-d" is to be interpreted as any sub-range comprised therein, e.g. d- . d- . d-d . d-d . d-d , d- ; particularly d-d.

Further, as used herein, the term "d-d", as used throughout this text, e.g. in the context of the definition of "d-d-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 "d-d" is to be interpreted as any sub-range comprised therein, e.g. d- , d-d , d- , d-d , d-d. d-d ; particularly d-d. The term "substituted" means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.

The term "optionally substituted" means optional substitution with the specified groups, radicals or moieties. As used herein, the term "one or more", e.g. in the definition of the substituents of the compounds of the general formulae of the present invention, is understood as meaning "one, two, three, four or five, particularly one, two, three or four, more particularly one, two or three, even more particularly one or two". As used herein, the term "leaving group" refers to an atom or a group of atoms that is displaced in a chemical reaction as stable species taking with it the bonding electrons.

Preferably, a leaving group is selected from the group comprising: halo, in particular chloro, bromo or iodo, methanesulfonyloxy, p-toluenesulfonyloxy, trifiuoromethanesulfonyloxy, nonafluorobutanesulfonyloxy, (4-bromo-benzene)sulfonyloxy, (4-nitro-benzene)sulfonyloxy, (2-nitro-benzene)-sulfonyloxy, (4-isopropyl-benzene)sulfonyloxy, (2,4.6-tri-isopropyl- benzene)-sulfonyloxy, (2.4,6-trimethyl-benzene)sulfonyloxy, (4-tertbutyl-benzene)sulfonyloxy, benzenesulfonyloxy, and (4-methoxy-benzene)sulfonyloxy.

As used herein, the term "protective group " is a protective group attached to a nitrogen in intermediates used for the preparation of compounds of the general formula (I). Such groups are introduced e.g. by chemical modification of the respective amino group in order to obtain chemoselectivity in a subsequent chemical reaction. Protective groups for amino groups are descibed for example in T.W. Greene and P.G.M. Wuts in Protective Groups in Organic Synthesis, 3 rd edition, Wiley 1999; more specifically, said groups can be selected from substituted sulfonyl groups, such as mesyl-, tosyl- or phenylsulfonyl-, acyl groups such as benzoyl, acetyl or tetrahydropyranoyl-, or carbamate based groups, such as tert.- butoxycarbonyl (Boc), or can include silicon, as in e.g. 2-(trimethylsilyl)ethoxymethyl (SEM).

The invention includes all suitable isotopic variations of a compound of the invention. An isotopic variation of a compound of the invention is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually or predominantly found in nature. Examples of isotopes that can be incorporated into a compound of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine, chlorine, bromine and iodine, such as 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, 8 F, 36 CI, 82 Br, 123 l, 124 l, 129 l and 3 1, respectively. Certain isotopic variations of a compound of the invention, for example, those in which one or more radioactive isotopes such as 3 H or 4 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 may be preferred in some circumstances. Isotopic variations of a compound of the invention can generally be prepared by conventional procedures known by a person skilled in the art such as by the illustrative methods or by the preparations described in the examples hereafter using appropriate isotopic variations of suitable reagents.

Where the plural form of the word compounds, salts, polymorphs, hydrates, solvates and the like, is used herein, this is taken to mean also a single compound, salt, polymorph, isomer, hydrate, solvate or the like. By "stable compound' or "stable structure" is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.

The compounds of this invention may contain one or more asymmetric centre, depending upon the location and nature of the various substituents desired. Asymmetric carbon atoms may be 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 of the present invention may contain sulphur atoms which are asymmetric, such as an asymmetric sulphoxide or sulphoximine group, 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 are those which produce the more desirable biological activity.

Separated, pure or partially purified isomers and stereoisomers or racemic or diastereomeric mixtures of the compounds of this invention are also included within the scope of the present invention. The purification and the separation of such materials can be accomplished by standard techniques known in the art. Pure stereoisomers can be obtained by resolution of 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 lUPAC Rules Section E (Pure Appl Chem 45, 1 1 -30, 1976).

The present invention includes all possible stereoisomers of the compounds of the present invention as single stereoisomers, or as any mixture of said stereoisomers, e.g. (R) or (S) isomers, or (£) or (Z) isomers, in any ratio. Isolation of a single stereoisomer, e.g. a single enantiomer or a single diastereomer, of a compound of the present invention may be achieved by any suitable state of the art method, such as chromatography, especially chiral chromatography, for example. Further, the compounds of the present invention may exist as tautomers. For example, any compound of the present invention which r~~* ; ~ " pyrazole moiety as a heteroaryl group for example can exist as a 1 H tautomer, or a 2H tautomer, or even a mixture in any amount of the two tautomers, or a triazole moiety for example can exist as a 1 H tautomer, a 2H tautomer, or a 4H tautomer, or even a mixture in any amount of said 1 H, 2H and 4H tautomers, namely :

1 H-tautomer 2H-tautomer 4H-tautomer

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

Further, the compounds of the present invention can exist as N -oxides, which are defined in that at least one nitrogen of the compounds of the present invention is oxidised. The present invention includes all such possible N -oxides. The present invention also relates to useful forms of the compounds as disclosed herein, such as metabolites, hydrates, solvates, prodrugs, salts, in particular pharmaceutically acceptable salts, and co-precipitates.

The compounds of the present invention can exist as a hydrate, or as a solvate, wherein the compounds of the present invention contain polar solvents, in particular water, methanol or ethanol for example as structural element of the crystal lattice of the compounds. The amount of polar solvents, in particular water, may exist in a stoichiometric or

non-stoichiometric ratio. In the case of stoichiometric solvates, e.g. a hydrate, hemi-, (semi-), mono-, sesqui-, di-, tri-, tetra-, penta- etc. solvates or hydrates, respectively, are possible. The present invention includes all such hydrates or solvates.

Further, the compounds of the present invention can exist in free form, e.g. as a free base, or as a free acid, or as a zwitterion, or can exist in the form of a salt. Said salt may be any salt, either an organic or inorganic addition salt, particularly any pharmaceutically acceptable organic or inorganic addition salt, customarily used in pharmacy.

The term "pharmaceutically acceptable salt" refers to a relatively non-toxic, inorganic or organic acid addition salt of a compound of the present invention. For example, see S. M. Berge, et al. "Pharmaceutical Salts, " J. Pharm. Sci. 1977, 66, 1 -19. A suitable pharmaceutically acceptable salt of the compounds of the present invention may be, for example, an acid-addition salt of a compound of the present invention bearing a nitrogen atom, in a chain or in a ring, for example, which is sufficiently basic, such as an acid-addition salt with an inorganic acid, such as hydrochloric, hydrobromic, hydroiodic, sulfuric, bisulfuric, phosphoric, or nitric acid, for example, or with an organic acid, such as formic, acetic, acetoacetic, pyruvic, trifluoroacetic, propionic, butyric, hexanoic, heptanoic, undecanoic, lauric, benzoic, salicylic, 2-(4-hydroxybenzoyl)-benzoic, camphoric, cinnamic, cyclopentanepropionic, digluconic, 3-hydroxy-2-naphthoic, nicotinic, pamoic, pectinic, persulfuric, 3-phenylpropionic, picric, pivalic, 2-hydroxyethanesulfonate, itaconic, sulfamic, trifluoromethanesulfonic, dodecylsulfuric, ethanesulfonic, benzenesulfonic,

para-toluenesulfonic, methanesulfonic, 2-naphthalenesulfonic, naphthalinedisulfonic, camphorsulfonic acid, citric, tartaric, stearic, lactic, oxalic, malonic, succinic, malic, adipic, alginic, maleic, fumaric, D-gluconic, mandelic, ascorbic, glucoheptanoic, glycerophosphoric, aspartic, sulfosalicylic, hemisulfuric, or thiocyanic acid, for example. Further, another suitably pharmaceutically acceptable salt of a compound of the present invention which is sufficiently acidic, is an alkali metal salt, for example a sodium or potassium salt, an alkaline earth metal salt, for example a calcium or magnesium salt, an ammonium salt or a salt with an organic base which affords a physiologically acceptable cation, for example a salt with N-methyl-glucamine, dimethyl-glucamine, ethyl-glucamine, lysine, dicyclohexylamine, 1 ,6-hexadiamine, ethanolamine, glucosamine, sarcosine, serinol, tris-hydroxy-methyl-aminomethane, aminopropandiol, sovak-base, 1-amino-2,3,4-butantriol, or with a quarternary ammonium salt, such as tetramethylammonium, tetraethylammonium, tetra(n-propyl)ammonium, tetra (n-butyl)ammonium, or /V-benzyl- /V,/V,A/-trimethylammonium.

Those skilled in the art will further recognise that acid addition salts of the claimed compounds may be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods. Alternatively, alkali and alkaline earth metal salts of acidic compounds of the invention are prepared by reacting the compounds of the invention with the appropriate base via a variety of known methods.

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

Furthermore, the present invention includes all possible crystalline forms, or polymorphs, of the compounds of the present invention, either as single polymorphs, or as a mixture of more than one polymorphs, in any ratio. In accordance with a first aspect, the present invention covers compounds of general formula

(I)

in which

represents a halogen atom or a group selected from:

hydroxy-, cyano-, Ci-Ce-alkyl-, Ci-Ce-alkoxy-, halo-Ci-Ce-alkyl-, halo-Ci

hydroxy-Ci-Ce-alkyl-, Ci-C 3 -alkoxy-Ci-C 3 -alkyl-, S-(Ci-C 3 -alkyl), represents a hydrogen atom or a group Ci-Ce-alkyl-;

R 3 represents a group selected from: -C(=0)-0-R 5 , -C(=0)-N(R 5a )R 5 ; R 4 represents halo-, hydroxy-, oxo- (0=), cyano-, nitro-, Ci-Ce-alkyl-, C2-Ce-alkenyl-, C2-Ce-alkynyl-, halo-Ci-Ce-alkyl-, Ci-Ce-alkoxy-, halo-Ci-Ce-alkoxy-,

hyd roxy-Ci -Ce-a I ky I-, Ci-Ce-alkoxy-Ci-Ce-alkyl-, halo-C -Ce-alkoxy-Ci-Ce-alkyk

R 6 -0-, -C(=0)-R 6 , -C(-0)-0-R 6 , -0-C(=0)-R e , -N(R 6a )-C(=0)-R 6b , -N(R 6a )-C(=0)-0- R 6b , -N(R 6a )-C(=0)-N(R 6b )R 6c , -N(R 6a )R 6b , -N(R 6a )R 6d , -C(-0)-N(R 6a )R 6b , R 6 -S-, R 6 -S(=0)-, R 6 -S(=0) 2 -, -N(R 6a )-S(=0)-R 6b , -S(=0)-N(R 6a )R 6b , -N(R 6a )-S(=0) 2 - R 6b , -S(=0) 2 -N(R 6a )R 6b , -S(=0)=N(R 6a )R 6b , -N=S(=0)(R 6a )R 6b or -(d-Ce-alkyl)- N(R 6a )R 6b ;

R 5 , R 5a , R 5b are the same or different and independently selected from each other, and represent a hydrogen atom or a group selected from: Ci-Ce-alkyl-, Cs-Ce-cycloalkyl-,

3- to 10-membered heterocycloalkyi-, aryl-, and heteroaryl-;

wherein said Ci-Ce-alkyl-, C3-Ce-cycloalkyl-, 3- to 10-membered heterocycloalkyi-, aryl- and heteroaryl- group is optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups ;or

N(R 5a )R 5 together represent a 3- to 10-membered heterocycloalkyi- group, said group being optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups ; R 6 , R 6a , R 6b , R 6c are the same or different and independently selected from each other, and represent a hydrogen atom, a d-Ce-alkyl- or a d-d-cycloalkyl- group: or

R 6a and R 6b , or

R 6a and R 6c , or

R 6b and R 6c together may form a C2-Ce-alkylene group, in which optionally one methylene can be replaced by -0-, -C(=0)-, -NH-, or -N(d-d-alkyl)- ;

R 6d represents -(d-d-alkyl)-N(R 6a )R 6b ; or a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.

In another preferred embodiment, the invention relates to compounds of formula (I), supra, wherein R represents a group selected from: cyano-, d-d-alkyl-, d-d-alkoxy-, halo-d-Ce-alkyl-, halo-d-d-alkoxy-, hydroxy-d-d-alkyl-, d-d-alkoxy-d-dralkyl-, and -S-(d-d-alkyl).

In another preferred embodiment, the invention relates to compounds of formula (I), supra, wherein R represents a group selected from: d-ds-alkyl-, d-d-alkoxy-, halo-d-d-alkyl-, halo-d-C 3 -alkoxy-, hydroxy-d-d,-alkyl-, d-d-alkoxy-d-d-alkyl-, and -S-(d-C 3 -alkyl).

In another preferred embodiment, the invention relates to compounds of formula (I), supra, wherein R 1 represents a group selected from: d-d,-alkyl-, d-dralkoxy-, halo-Ci-C 3 -alkyl-, and -S-(Ci-C 3 -alkyl). In another preferred embodiment, the invention relates to compounds of formula (I), supra, wherein R 1 represents a d-d-alkoxy-.

In another preferred embodiment, the invention relates to compounds of formula (I), supra, wherein R 1 represents a chlorine atom or florine atom or a group -0-CH 3 .

In another preferred embodiment, the invention relates to compounds of formula (I), supra, wherein R 1 represents a chlorine atom or a -0-CH 3 group.

In another preferred embodiment, the invention relates to compounds of formula (I), supra, wherein R represents a chlorine atom.

In another preferred embodiment, the invention relates to compounds of formula (I), supra, wherein R 1 represents a -O-CH3 group. In another preferred embodiment, the invention relates to compounds of formula (I), wherein R 2 represents a hydrogen atom.

In another preferred embodiment, the invention relates to com ounds of formula (la):

(la) in which R 1 , R a and R 5 are as defined for compounds of formula (I).

In another preferred embodiment, the invention relates to compounds of formula (la) with defined sterochemstry:

(la) in which R\ R 5a and R 5 are as defined for compounds of formula (I).

In another preferred embodiment, the invention relates to compounds of formula (la), supra, in which R 1 represents a -O-CH3 group , and in which R 5a and R 5 are as defined for compounds of formula (I).

In another preferred embodiment, the invention relates to compounds of formula (la), supra, in which R 1 represents a chlorine atom, and in which R 5a and R 5 are as defined for compounds of formula (I).

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents halo-, hydroxy-, cyano-, nitro-, Ci-Ce-alkyl-, halo-Ci-Ce-alkyl-, Ci-Ce-alkoxy-, halo-Ci-Ce-alkoxy-, hydroxy-d-Ce-alkyl-, Ci-Ce-alkoxy-Ci-Ce-alkyl-, h a lo-C -Ce-a I koxy-C -Ce-a I ky I- , R 6 -0-, -C(=0)-R 6 , -C(=0)-0-R 6 , -0-C(=0)-R 6 , -N(R 6a )-C(=0)- R 6b , -N(R 6a )-C(=0)-N(R 6b )R 6c , -N(R 6a )R 6b , -C(=0)-N(R 6a )R 6b , R 6 -S-, R 6 -S(=0)-,

R e -S(=0) 2 -, -N(R 6a )-S(=0)-R 6b , -S(=0)-N(R 6a )R 6b , -N(R 6a )-S(=0) 2 - R 6b , -S(=0) 2 -N(R 6a )R 6b , -S(=0)=N(R 6a )R 6b or -N=S(=0)(R 6a )R 6b .

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents halo-, hydroxy-, cyano-, nitro-, Ci-Ca-alkyl-, halo-Ci-Cs-alkyl-, Ci-Cs-alkoxy-, halo-Ci-Cs-alkoxy-, hyd roxy-Ci -Cs-a I kyl-, Ci-Cr-alkoxy-Ci-Cs-alkyl-, h a lo-C -C 3 -a I koxy-C -C 3 -a I ky I- , R 6 -0-, -C(=0)-R 6 , -C(=0)-0-R 6 , -0-C(=0)-R 6 , -

N(R 6a )R 6b , -C(=0)-N(R 6a )R eb , R 6 -S-, R 6 -S(=0)-, R e -S(=0) 2 -, -N(R 6a )-S(=0)-R 6b , -S(=0)- N(R 6a )R eb , -N(R 6a )-S(=0) 2 -R 6b , or -S(=0) 2 -N(R 6a )R 6b .

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents halo-, hydroxy-, cyano-, Ci-Cs-alkyl-, halo-d-Cs-alkyl-, C -Cs-alkoxy-, halo-Ci-Cs-alkoxy-, hyd roxy-C -Cs-a I kyl-, Ci-Cr-alkoxy-Ci-Cs-alkyl-, or -

N(R 6a )R 6b .

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents halo-, hydroxy-, cyano-, Ci-Ce-alkyl-, Ci-Ce-alkoxy-,

-N(R 6a )-C(=0)-0-R 6b , -N(R 6a )R 6b , -N(R 6a )R 6d , -C(=0)-N(R 6a )R 6b or -(Ci-Ce-alkyl)-N(R 6a )R 6b .

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents halo-, hydroxy-, cyano-, Ci-C:,-alkyl-, Ci-Cs-alkoxy-,

-N(R 6a )-C(=0)-0-R 6b , -N(R 6a )R 6b , -N(R 6a )R 6d , -C(=0)-N(R 6a )R 6b or -(CrC 3 -alkyl)-N(R 6a )R 6b .

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents hydroxy-, Ci-Cs-alkyl-, -N(R 6a )R 6b or -N(R 6a )R 6d . In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents hydroxy-, Ci-Cs-alkyl-, -N(R 6a )R 6b or -N(R 6a )R 6d , and in which R 1 represents a hydrogen atom. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents hydroxy-, Ci-C 3 -alkyl-, -N(R 6a )R 6b or -N(R 6a )R 6d , and in which R 2 represents a hydrogen atom.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents hydroxy-, Ci-Cs-alkyl-, -N(R 6a )R 6b or -N(R 6a )R 6d , and in which R 1 represents a hydrogen atom and R 2 represents a hydrogen atom. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents fluoro-, hydroxy-, cyano-, methyl-, methoxy-, - (CH3)2, -NH2.

-(Ci-C 3 -alkyl)-N(CH 3 )2, -C(=0)-N(CH 3 ) 2 , -N(CH 3 )-(Ci-C 3 -alkyl)-N(CH 3 ) 2 or

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents fluoro-, hydroxy-, cyano-, methyl-, methoxy-, -N(CH 3 )2, -NH2, -(Ci-C 3 -alkyl)-N(CH 3 ) 2 , -C(=0)-N(CH 3 ) 2 , -N(CH 3 )-(Ci-C 3 -alkyl)-N(CH 3 ) 2 or

-N(H)-C(=0)-0-(Ci-C4-alkyl), and in which R 1 represents a hydrogen atom. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents fluoro-, hydroxy-, cyano-, methyl-, methoxy-, -N(CH 3 )2, -NH2. -(Ci-C 3 -alkyl)-N(CH 3 )2, -C(=0)-N(CH 3 ) 2 , -N(CH 3 )-(Ci-C 3 -alkyl)-N(CH 3 ) 2 or

-N(H)-C(=0)-0-(Ci-C4-alkyl), and in which R 2 represents a hydrogen atom. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents fluoro-, hydroxy-, cyano-, methyl-, methoxy-, -N(CH 3 ) 2 , -NH 2 , -(Ci-C 3 -alkyl)-N(CH 3 ) 2 , -C(=0)-N(CH 3 ) 2 , -N(CH 3 )-(C,-C 3 -alkyl)-N(CH 3 ) 2 or

-N(H)-C(=0)-0-(Ci-C4-alkyl), and in which R 1 represents a hydrogen atom and R 2 represents a hydrogen atom.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents hydroxy-, methyl-, -N(CH 3 ) 2 , -NH2 or -N(CH 3 )-(Ci-C 3 -alkyl)- N(CH 3 ) 2 . In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents hydroxy-, methyl-, -N(CH 3 ) 2 , -NH 2 or -N(CH 3 )-(CH 2 ) 2 -N(CH 3 ) 2 . In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents halo-, hydroxy-, cyano-, Ci-Cs-alkyk Ci-Cs-alkoxy-, -N(R 6a )- C(=0)-0-, -N(R 6a )R 6b or -C(=0)-N(R 6a )R 6b . In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents fluoro-, hydroxy-, cyano-, C-Cs-alkyl-, Ci-Cs-alkoxy-, -N(R 6a )- C(=0)-0-, -N(R 6a )R 6b or -C(=0)-N(R 6a )R 6b .

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents halo-, hydroxy-, cyano-, Ci-C:,-alkyl-, C-C:,-alkoxy-, or -

N(R 6a )R 6b .

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 4 represents fluoro-, hydroxy-, cyano-, Ci-Cs-alkyl-, C-Cs-alkoxy-, or - N(R 6a )R 6b .

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a hydrogen atom. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a Ci-Ce-alkyl- group: wherein said Ci-Ce-alkyl- group is optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a C -C3-alkyl- group; wherein said Ci-C:,-alkyl- group is optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a Ci-C:,-alkyl- group; wherein said Ci-Cs-alkyl- group is optionally substituted, identically or differently, with 1 or 2 R 4 groups.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a Ci-Cs-alkyl- group, wherein said Ci-Cs-alkyl- group is substituted with a -N(R 6a )R 6b group. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a C 2 -C3-alkyl- group, wherein said C2-C3-alkyl- group is substituted with a -N(CH 3 )2 or -NH 2 group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a -(CH 2 )2-NH 2 group or a -(CH 2 )3-N(CH 3 )2 group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a Ci-Cr-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a 3- to 10-membered heterocycloalkyi- group, wherein said 3- to 10-membered heterocycloalkyi- group is optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a group selected from: Ci-Ci-alkyl-. 4- to 7-membered heterocycloalkyi-; wherein said Ci-Ci-alkyl- group and 4- to 7-membered heterocycloalkyl- group are optionally substituted, identically or differently, with 1 or 2 R 4 groups.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a group selected from: Ci-Cj-alkyl-, 4- to 7-membered heterocycloalkyi-; wherein said 4- to 7-membered heterocycloalkyi- group is selected from: oxaazaspiro[3.3]heptyl, azabicyclo[3.1 OJhexyl, oxazabicyclo[2.2.1 jheptyl-,

diazabicyclo[2.2.1 Jheptyl, pyrrolidinyl, piperidinyl, piperazinyl, azetidinyl, morpholinyl;

wherein said Ci-Ci-alkyl- group and 4- to 7-membered heterocycloalkyl- group are optionally substituted, identically or differently, with 1 or 2 R 4 groups.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a group selected from: Ci-Ci-alkyl-, 4- to 7-membered heterocycloalkyi-; wherein said 4- to 7-membered heterocycloalkyi- group is selected from: oxaazaspiro[3.3]heptyl, azabicyclo[3.1 Ojhexyl, oxazabicyclo[2.2.1 jheptyl-,

diazabicyclo[2.2.1 Jheptyl, pyrrolidinyl, piperidinyl, piperazinyl, azetidinyl, morpholinyl;

wherein said Ci-C4-alkyl- group and 4- to 7-membered heterocycloalkyl- group are optionally substituted, identically or differently, with 1 or 2 R 4 groups, and in which compounds R 1 represents a hydrogen atom. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a group selected from: Ci-Gs-alkyl-, 4- to 7-membered heterocycloalkyl-; wherein said 4- to 7-membered heterocycloalkyl- group is selected from: oxaazaspiro[3.3]heptyl, azabicyclo[3.1 .OJhexyl, oxazabicyclo[2.2.1 Jheptyl-,

diazabicyclo[2.2.1 Jheptyl, pyrrolidinyl, piperidinyl, piperazinyl, azetidinyl, morpholinyl;

wherein said C -C4-alkyl- group and 4- to 7-membered heterocycloalkyl- group are optionally substituted, identically or differently, with 1 or 2 R 4 groups, and in which compounds R 2 represents a hydrogen atom. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a group selected from: Ci-Ci-alkyl-, 4- to 7-membered heterocycloalkyl-; wherein said 4- to 7-membered heterocycloalkyl- group is selected from: oxaazaspiro[3.3]heptyl, azabicyclo[3.1 .Ojhexyl, oxazabicyclo[2.2.1 Jheptyl-,

diazabicyclo[2.2.1 Jheptyl, pyrrolidinyl, piperidinyl, piperazinyl, azetidinyl, morpholinyl;

wherein said Ci-Ci-alkyl- group and 4- to 7-membered heterocycloalkyl- group are optionally substituted, identically or differently, with 1 or 2 R 4 groups, and in which compounds R 1 represents a hydrogen atom, and R 2 represents a hydrogen atom.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5 represents a group selected from: C2-Cs-alkyl-, 4- to 7-membered heterocycloalkyl-; wherein said 4- to 7-membered heterocycloalkyl- group is selected from: diazabicyclo[2.2.1 Jheptyl, pyrrolidinyl, piperidinyl, azetidinyl;

wherein said C2-C3-alkyl- group and 4- to 7-membered heterocycloalkyl- group are optionally substituted, identically or differently, with 1 or 2 R 4 groups.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5a represents a hydrogen atom or a Ci-Cs-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5a represents a hydrogen atom.

In another preferred embodiment, the invention relates to compounds of formula (I), supra, wherein R 5a represents a Ci-Ce-alkyl- group; wherein said d-Ce-alkyl- group is optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein 5a represents a Ci-Cs-alkyl- group; wherein said Ci-Cs-alkyl- group is optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5a represents a Ci-Cr-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5a represents a methyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5b represents a hydrogen atom or a Ci-Cs-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5b represents a hydrogen atom.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5b represents a Ci-Ce-alkyl- group; wherein said C -Ce-alkyl- group is optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5b represents a Ci-Cr-alkyl- group; wherein said C -Cs-alkyl- group is optionally substituted, identically or differently, with 1 . 2, 3. 4 or 5 R 4 groups. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5b represents a Ci-Cs-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5b represents a methyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 5b represents a 3- to 10-membered heterocycloalkyi- group, wherein said 3- to 10-membered heterocycloalkyi- group is optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R b represents a 4- to 7-mr~' ' heterocycloalkyi- group, wherein said 4- to 7-membered heterocycloaikyi- group is optionally substituted, identically or differently, with 1 , 2 or 3 4 groups.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein N(R 5a )R 5 together represent a 3- to 10-membered heterocycloaikyi- group, said group being optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein N(R 5a )R 5 together represent a 4- to 7-membered heterocycloaikyi- group, said group being optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein N(R 5a )R 5 together represent a 4- to 7-membered heterocycloaikyi- group, wherein said 4- to 7-membered heterocycloaikyi- group is selected from:

oxaazaspiro[3.3]heptyl, azabicyclo[3.1 .OJhexyl, oxazabicyclo[2.2.1 Jheptyl-,

diazabicyclo[2.2.1 ]heptyl, pyrrolidinyl, piperidinyl, piperazinyl, azetidinyl, morpholinyl;

wherein said 4- to 7-membered heterocycloaikyi- group is optionally substituted, identically or differently, with 1 or 2 R 4 groups. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein N(R 5a )R 5 together represent a 4- to 7-membered heterocycloaikyi- group, wherein said 4- to 7-membered heterocycloaikyi- group is selected from:

oxaazaspiro[3.3]heptyl, azabicyclo[3.1 .OJhexyl, oxazabicyclo[2.2.1 ]heptyl-,

diazabicyclo[2.2.1 Jheptyl, pyrrolidinyl, piperidinyl, piperazinyl, azetidinyl, morpholinyl:

wherein said 4- to 7-membered heterocycloaikyi- group is optionally substituted, identically or differently, with 1 or 2 R 4 groups, and in which compounds R 1 represents a hydrogen atom.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein N(R 5a )R 5 together represent a 4- to 7-membered heterocycloaikyi- group, wherein said 4- to 7-membered heterocycloaikyi- group is selected from:

oxaazaspiro[3.3]heptyl, azabicyclo[3.1 .0]hexyl, oxazabicyclo[2.2.1 Jheptyl-,

diazabicyclo[2.2.1 Jheptyl, pyrrolidinyl, piperidinyl, piperazinyl, azetidinyl, morpholinyl;

wherein said 4- to 7-membered heterocycloaikyi- group is optionally substituted, identically or differently, with 1 or 2 R 4 groups, and in which compounds R 2 represents a hydrogen atom.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein N(R 5a )R 5 together represent a 4- to 7-membered heterocycloaikyi- group, wherein said 4- to 7-membered heterocycloaikyi- group is selected from:

oxaazaspiro[3.3]heptyl, azabicyclo[3.1 .Ojhexyl, oxazabicyclo[2.2.1 ]heptyl-,

diazabicyclo[2.2.1 Jheptyl. pyrrolidinyl, piperidinyl, piperazinyl, azetidinyl, morpholinyl;

wherein said 4- to 7-membered heterocycloaikyi- group is optionally substituted, identically or differently, with 1 or 2 R 4 groups, and in which compounds R 1 represents a hydrogen atom, and R 2 represents a hydrogen atom.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein N(R 5a )R 5 together represent a 4- to 7-membered heterocycloaikyi- group, wherein said 4- to 7-membered heterocycloaikyi- group is selected from:

diazabicyclo[2.2.1 Jheptyl, pyrrolidinyl, piperidinyl, azetidinyl;

wherein said 4- to 7-membered heterocycloaikyi- group is optionally substituted, identically or differently, with 1 or 2 R 4 groups. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein N(R 5a )R 5 together represent a group selected from:

wherein * indicates the point of attachment of said groups with the rest of the molecule.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein N(R 5a ) 5 together represent a group selected from:

wherein * indicates the point of attachment of said groups with the rest of the molecule. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6 represents a hydrogen atom or a Ci-Ce-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6 represents a hydrogen atom or a Ci-Cs-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6 represents a Ci-C:,-alkyl- group. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6 represents a methyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6a represents a hydrogen atom or a Ci-Ce-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6a represents a hydrogen atom or a d-Cs-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6a represents a Ci-Cr-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6a represents a methyl- group. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6a represents a methyl- group and wherein R 6b represents a methyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6a represents a methyl- group, wherein R 6b represents a methyl- group, and wherein R 2 represents a hydrogen atom.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6a represents a methyl- group, wherein R 6b represents a methyl- group, and wherein R 1 represents a hydrogen atom. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6a represents a methyl- group, wherein R 6b represents a methyl- group, and wherein R 1 represents a -O-CH3 group. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6a represents a methyl- group, wherein R 6b represents a methyl- group, wherein R 2 represents a hydrogen atom, and wherein R 1 represents a -O-CH3 group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6b represents a hydrogen atom or a Ci-Ce-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6b represents a hydrogen atom or a C -Cs-alkyl- group. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6b represents a C -Cs-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6b represents a methyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6c represents a hydrogen atom or a Ci-Ce-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6c represents a hydrogen atom or a Ci-C:,-alkyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6c represents a Ci-Cr-alkyl- group. In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6c represents a methyl- group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6d represents a -(Ci-Cs-alkyl)-N(R 6a )R 6b group.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, wherein R 6d represents a -(Ci-C3-alkyl)-N(CH 3 )2 group. It is to be understood that the present invention relates also to any combination of the preferred embodiments described above. Some examples of combinations are given hereinafter. However, the invention is not limited to these combinations.

In a preferred embodiment, the invention relates to compounds of formula (I), supra, in which :

R 1 represents a halogen atom or a group selected from: Ci-C:,-alkyl-, Ci-Cs-alkoxy-, halo-d-Cs-alkyl-, and -S-(Ci-C 3 -alkyl);

R 2 represents a hydrogen atom;

R 3 represents a group -C(=0)-N(R 5a )R 5 ; R 4 represents halo-, hydroxy-, cyano-, nitro-, Ci-Cs-alkyl-, halo-Ci-Cs-alkyk

Ci-C:,-alkoxy-, halo-Ci-Cs-alkoxy-, hydroxy-Ci-C:,-alkyl-, Ci-Cs-alkoxy-Ci-Cs-alkyl-, halo-C-Cs-alkoxy-Ci-Cs-alkyl-, R 6 -0-, -C(=0)-R 6 , -C(=0)-0-R 6 , -0-C(=0)-R 6 ,

-N(R 6a )R 6b , -C(=0)-N(R 6a )R 6b , R e -S-, R e -S(=0)-, R e -S(=0) 2 -, -N(R 6a )-S(=0)- R 6b , -S(=0)-N(R 6a )R 6b , -N(R 6a )-S(=0) 2 -R 6b , or -S(=0) 2 -N(R 6a )R 6b ;

R 5 , R 5a , R 5b are the same or different and independently selected from each other, and represent a hydrogen atom or a group selected from: d-Ce-alkyl-, Cs-Ce-cycloalkyl-, 3- to 10-membered heterocycloalkyi-, aryk and heteroaryl-;

wherein said Ci-Ce-alkyl-, Cr-Ce-cycloalkyl-, 3- to 10-membered heterocycloalkyi-, aryl- and heteroaryl- group is optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups; or

N(R 5a )R 5 together represent a 3- to 10-membered heterocycloalkyi- group, said group being optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups ;

R 6a , R 6b , R 6c are the same or different and are independently selected from R 6 ;

R 6 , R 6a , R 6b , R 6c are the same or different and independently selected from each other, and represent a hydrogen atom or a Ci-Ce-alkyl- group;

or a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.

In another preferred embodiment, the invention relates to compounds of formula (I), supra, in which : R 1 represents a group selected from: Ci-C:,-alkyl-, Ci-Ca-alkoxy-, halo-Ci-C:,-alkyl-, and S-(Ci-C 3 -alkyl);

R 2 represents a hydrogen atom;

R 3 represents a group -C(=0)-N(R 5a )R 5 ;

R 4 represents halo-, hydroxy-, cyano-, Ci-Cs-alkyl-, halo-Ci-C:,-alkyl-, Ci-Cs-alkoxy-, halo-Ci-C 3 -alkoxy-, hydroxy-Ci-C 3 -alkyl-, Ci-Cr-alkoxy-Ci-Cs-alkyl-, -NR 6a R 6b ,

-(Ci-Ce-alkyl)-N(R ea )R 6b or -N(R 6a )R 6d ;

R 5 , R 5a , R 5b are the same or different and independently selected from each other, and represent a hydrogen atom or a d-Cs-alkyl- group;

wherein said Ci-Cs-alkyl- group is optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups;or

N(R 5a )R 5 together represent a 3- to 10-membered heterocycloalkyi- group, said group being optionally substituted, identically or differently, with 1 or 2 R 4 groups ;

R 6 , R 6a , R 6b , R 6c are the same or different and independently selected from each other, and represents a hydrogen atom or a Ci-Ce-alkyl- group ;

R 6d represents a -(Ci-C 3 -alkyl)-NR 6a R 6b group ;

or a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.

In another preferred embodiment, the invention relates to compounds of formula (I), supra, in which :

R 1 represents a group selected from: Ci-C:-alkyk Ci-C:,-alkoxy-, halo-Ci-Cs-alkyl-, and - S-(Ci-C 3 -alkyl);

R 2 represents a hydrogen atom;

R 3 represents a group -C(=0)-N(R 5a )R 5 ;

R 4 represents halo-, hydroxy-, cyano-, Ci-Cs-alkyk halo-Ci-Cs-alkyl-, Ci-C:-alkoxy-, halo-Ci-Cs-alkoxy-, hydroxy-Ci-Cs-alkyl-, Ci-Cs-alkoxy-C -Cs-alkyl-, or -N(R 6a )R 6b ;

R 5 , R 5a , R 5b are the same or different and independently selected from each other, and represent a hydrogen atom or a Ci-C:,-alkyl- group; wherein said Ci-C 3 -alkyl- group is optionally substituted, identically or differently, with 1 , 2, 3, 4 or 5 R 4 groups; or

N(R 5a )R 5 together represent a 3- to 10-membered heterocycloalkyi- group, said group being optionally substituted, identically or differently, with 1 or 2 R 4 groups ;

R 6 , R 6a , R 6b , R 6c are the same or different and independently selected from each other, and represent a hydrogen atom or a C-Ce-alkyl- group ;

or a tautomer, an -oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.

In another preferred embodiment, the invention relates to compounds of formula (I), supra, in which :

R represents a chlorine atom or a group -O-CH3; R 2 represents a hydrogen atom; R 3 represents -C(=0)-N(R 5a )R 5 ;

R 4 represents halo-, hydroxy-, cyano-, Ci-Cs-alkyl-, halo-Ci-Cs-alkyl-, Ci-Cs-alkoxy-, halo-C-C 3 -alkoxy-, hydroxy-Ci-C 3 -alkyl-, Ci-C 3 -alkoxy-Ci-C 3 -alkyh or -N(R 6a )R 6b ;

R 5 , R 5a , R 5b are the same or different and independently selected from each other, and represent a hydrogen atom or a Ci-C 3 -alkyl- group;

wherein said C-C 3 -alkyl- group is optionally substituted, identically or differently, with 1 or 2 R 4 groups; or

N(R 5a )R 5 together represent a 3- to 10-membered heterocycloalkyi- group, said group being optionally substituted, identically or differently, with 1 or 2 R 4 groups ;

R 6 , R 6a , R 6b , R 6c are the same or different and independently selected from each other, and represent a hydrogen atom or a Ci-C 3 -alkyl- group ;

or a tautomer, an N-oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.

In another preferred embodiment, the invention relates to compounds of formula (I), supra, in which :

R 1 represents a chlorine atom or a group -0-CH 3 ;

R 2 represents a hydrogen atom R 3 represents -C(=0)-N(R 5a )R 5 ;

R 4 represents halo-, hydroxy-, cyano-, Ci-C 3 -alkyl-, Ci-C 3 -alkoxy-, -N(R 6a )-C(=0)-0-R 6 ~N(R 6a )R 6b , -N(R 6a )R 6d , -C(=0)-N(R 6a )R 6b or -(Ci-C 3 -alkyl)-N(R 6a )R 6b ;

R 5 represents a group selected from: C -Cs-alkyl-, 4- to 7-membered heterocycloalkyi-; wherein said Ci-Ci-alkyl- group and 4- to 7-membered heterocycloalkyi- group are optionally substituted, identically or differently, with 1 or 2 R 4 groups ;

R 5a represents a Ci-C 3 -alkyl- group ; or

N(R 5a )R 5 together represent a 4- to 7-membered heterocycloalkyi- group, said group being optionally substituted, identically or differently, with 1 or 2 R 4 groups ;

R 6a , R 6b are the same or different and are independently selected from R 6 ;

R 6 , R 6a , R 6b , R 6c are the same or different and independently selected from each other, and represent a hydrogen atom or a Ci-C- 3 -alkyl- group ;

R 6d represents a -(Ci-C 3 -alkyl)-N(R 6a )R 6b group ;

or a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.

In another preferred embodiment, the invention relates to compounds of formula (I) or (lb), supra, in which :

R 1 represents a chlorine atom or a group -0-CH 3 ; represents a hydrogen atom ; represents a -C(=0)-N(R 5a )R 5

R 4 represents halo-, hydroxy-, cyano-, Ci-C 3 -alkyl-, Ci-C 3 -alkoxy-, -N(R 6a )-C(=0)-0-R' -N(R 6a )R 6b , -N(R 6a )R 6d , -C(=0)-N(R 6a )R 6b or -(Ci-C 3 -alkyl)-N(R 6a )R 6b ; R 5 represents a group selected from: Ci-Gj-alkyl-;

wherein said C -C-!-alkyl- group is optionally substituted, identically or differently, with 1 or 2 R 4 groups ;

R 5a represents a Ci-C 3 -alkyl- group; or

N(R 5a )R 5 together represent a 4- to 7-membered heterocycloalkyi- group;

wherein said 4- to 7-membered heterocycloalkyi- group is selected from:

oxaazaspiro[3.3]heptyl, azabicyclo[3.1 .Ojhexyl, oxazabicyclo[2.2.1 Jheptyl-, diazabicyclo[2.2.1 Jheptyl, pyrrol id inyl, piperidinyl, piperazinyl, azetidinyl, morpholinyl; wherein said 4- to 7-membered heterocycloalkyi- group is optionally substituted, identically or differently, with 1 or 2 R 4 groups;

R 6 , R 6a , R 6b , R 6c are the same or different and independently selected from each other, and represents a hydrogen atom or a C -Cs-alkyl- group ;

R 6d represents a -(Ci-C 3 -alkyl)-N(R 6a )R 6b group ; or a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.

In another preferred embodiment, the invention relates to compounds of formula (I) or (la), supra, in which :

R 1 represents a chlorine atom or a group -O-CH-,; R 2 represents a hydrogen atom ;

R 3 represents a -C(=0)-N(R 5a )R 5 ;

R 4 represents hydroxy-, Ci-Cs-alkyl-, -N(R 6a )R 6b or -N(R 6a )R 6d ;

R 5a represents a methyl- group ;

R 5 represents a group selected from: Ci-Cs-alkyl-;

wherein said Ci-Ca-alkyl- group is substituted with a -N(R 6a )R 6b group ; or

N(R 5a )R 5 together represent a 4- to 7-membered heterocycloalkyi- group;

wherein said 4- to 7-membered heterocycloalkyi- group is selected from:

diazabicyclo[2.2.1 ]heptyl, pyrrolidinyl, piperidinyl, azetidinyl;

wherein said 4- to 7-membered heterocycloalkyi- group is optionally substituted, identically or differently, with 1 or 2 R 4 groups;

R 6b are the same or different and are independently selected from R 6 ;

represents a hydrogen atom or a methyl- group ; represents a -(Ci-C3-alkyl)-N(CH 3 )2 group or a tautomer. an N -oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same.

In another preferred embodiment, the invention relates to com ounds of formula (la):

(la) in which :

R 1 represents a chlorine atom or a methoxy- group ; R 4 represents fluoro-, hydroxy-, cyano-, methyl-, methoxy-, -N(R 6a )-C(=0)-0-R f

-N(R 6a )R 6b or -C(=0)-N(R 6a )R (

R 5a represents a hydrogen atom or a methyl group; R 5 represents a d-Ce-alkyl- group;

wherein said C -Ce-alkyl- group is optionally substituted, identically or differently, with

1 , 2 or 3 R 4 groups; or

N(R 5a )R 5 together represent a 4- to 7-membered heterocycloaikyi- group, said group being optionally substituted, identically or differently, with 1 or 2 R 4 groups ;

R 6a represents a hydrogen atom or a methyl- group; R 6b represents a hydrogen atom or a methyl- group; or a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, or a mixture of same. It is to be understood that the present invention relates to any sub-combination within any embodiment or aspect of the present invention of compounds of general formula (I), supra.

More particularly still, the present invention covers compounds of general formula (I) which are disclosed in the Examples section of this text, infra.

In accordance with another aspect, the present invention covers methods of preparing compounds of the present invention, said methods comprising the steps as described in the Experimental Section herein.

In a preferred embodiment, the present invention relates to a method of preparing compounds of general formula (I), supra, in which method an intermediate compound of general formula (II):

(I I)

in which R 2 and R 3 are as defined for general formula (I), supra, and LG represents a leaving group, preferably a chlorine atom;

is allowed to react with an intermediate com ound of general formula (III):

(ill)

in which R is as defined for general formula (I), supra.

In accordance with a further aspect, the present invention covers intermediate compounds which are useful in the preparation of compounds of the present invention of general formula (I), particularly in the method described herein. In particular, the present invention covers compounds of general formula (II):

in which R 2 and R 3 are as defined for the compounds of general formula (I), supra, and LG represents a leaving group, preferably a chlorine atom.

Synthesis of compounds of general formula (I) of the present invention

Compounds of general formula (I) can be synthesized according to the general p raced depicted in Scheme 1 , wherein LG stands for a leaving group.

Scheme 1

Scheme 1 exemplifies the main route that allows variations in R\ R 2 and R 3 . The coupling of compounds of formula (II) with pyrazolo[1 ,5-a]pyridin-5-amines such as (III) can be accomplished by reacting the two reactants in a suitable solvent, such as ethanol or a related lower aliphatic alcohol of the formula Ci-C4-alkyl-OH or a cyclic ether, such as

tetrahydrofuran or 1 ,4-dioxane, optionally in the presence of an acid such as hydrochloric acid. The compounds of formula (III) can be used either as free base or as corresponding salt with organic or inorganic acids. Alternatively, such amination reactions can be performed using catalysis by metals, such as palladium (see e.g. J. Y. Yoon et al., Synthesis 2009, (5), 815, and literature cited therein), to give compounds of formulae (I) or (III).

Modification of any of the substituents, R 1 , R 2 and R 3 can be achieved before and/or after the exemplified transformation. However, also other routes may be used to synthesise the target compounds, in accordance with common general knowledge of a person skilled in the art of organic synthesis. Said modifications can be such as the introduction of protecting groups, cleavage of protecting groups, reduction or oxidation of functional groups, formation or cleavage of esters or carboxamides, halogenation, metallation, substitution or other reactions known to a person skilled in the art. These transformations include those which introduce a functionality which allows for further interconversion of substituents. Appropriate protecting groups and their introduction and cleavage are well-known to a person skilled in the art (see for example T.W. Greene and P.G.M. Wuts in Protective Groups in Organic Synthesis, 3 rd edition, Wiley 1999). Further, it is possible that two or more successive steps may be performed without work-up being performed between said steps, e.g. a "one-pot" reaction, as it is well-known to a person skilled in the art.

Compounds of the general formula (II), wherein R 2 and R 3 have the meanings as given for general formula (I), and wherein LG stands for a leaving group, can be readily prepared as shown in Scheme 2 by a so-called Gewald thiophene synthesis (for a seminal publication see e.g. K. Gewald ef al., Chem. Ber. 1966, 94, 99), starting from ketones of the general formula (IV), to give the intermediate thiophene derivatives (V). Said intermediates are then cyclised to the thienopyrimidones (VI) employing a suitable Ci synthon such as formamide. The resulting pyrimidones (VI) are then transferred into compounds of the general formula (II) by suitable procedures known to the person skilled in the art, such as treatment with a chlorinating agent. An instructive exemplary protocol for the sequence outlined in Scheme 2 can be found in WO 2005/010008, example 14, steps 1 to 3.

If R 3 in compounds of the formula (II) comprises a carboxylic ester, e.g. an ethyl ester, it is well possible to convert said ester into a carboxamide in the presence of LG e.g.

representing a chloride, by mild ester hydrolysis using e.g. lithium hydroxide, followed by carboxamide coupling by procedures well known to the person skilled in the art.

Scheme 2

(iv) (V) (VI)

Multiple methods of isolating pure enantiomers from isomeric mixtures, e.g. racemic mixtures of chiral compounds are known to the person skilled in the art. Said methods encompass preparative HPLC on chiral stationary phase, kinetic resolution of racemic mixtures (for some examples see e.g. I. Shiina et al., Catal. Sci Technol. 2012, 2, 2200-2205; I. Shiina et al., Eur. J. Org. Chem. 2008. 5887-5890; D. G. Walker et al., Organic Process Research & Development 2001 , 5, 23-27; B. N. Roy et al., Organic Process Research & Development 2009, 13, 450; T. Storz and P. Dittmar, Organic Process Research & Development 2003, 7, 559), enantioselective protonation (for some examples see e.g. C. Fehr and G. Galindo, Helv. Chim. Acta 1995, 78, 539-552, S. Hunig et al.. Chem. Ber. 1994, 127, 1981 -1988; S. Hunig et al., Chem. Ber. 1994, 127, 1969), enzymatic resolution (for some examples see e.g. T. Miyazawa, Amino Acids 1999, 16. 191 -213), or, preferably and outlined in more detail below, temporary derivatisation with an enantiopure chiral synthon, separation of the resulting diastereomers, and removal of said chiral synthon, resulting in the isolation of the pure enantiomers of the parent compound (for some examples see e.g. Asymmetric

Synthesis - The Essentials. Edited by Mathias Christmann and Stefan Brase WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim).

Scheme 3

Scheme 3 illustrates the transformation of racemic pyrimidine synthons of the formula (lla- rac), in which R E represents a d-Ce-alkyl group, and in which Y stands for a leaving group LG or a hydroxyl group, into an activated form such as an acid chloride of the formula (VII- rac). It is well possible to hydrolyse the ester group present in said synthons (lla-rac) in the presence of Y e.g. representing a group LG e.g. representing a chloride, by mild ester hydrolysis using e.g. lithium hydroxide, as known by the person skilled in the art, to give carboxyiic acids of formula (llb-rac). These can be readily converted into acid chlorides of the formula (Vll-rac) by methods well known to the person skilled in the art, such as the reaction with an inorganic acid chloride such as thionyl chloride.

Said acid chlorides (Vll-rac) are subsequently reacted with a chiral, enantiomerically pure synthon such as an oxazolidinone of the formula (VIII), in which R 0x1 represents a hydrogen atom or a Ci-C<i-alkyl group, preferably methyl, and in which R 0x2 represents an aryl, aryl- (CH2)n- or a Ci-C-i-alkyl- group, wherein n is an integer selected from 1 , 2 and 3, and wherein R 0x2 preferably represents phenyl, after deprotonation of said oxazolidinone using a suitable deprotonation agent such as n-butyllithium or sodium hydride, at temperatures ranging from - 78°C to 0°C, preferably below -40°C, to give the amide coupling product of formula (IX) as mixture of two diastereoisomers. Said mixture can then be separated into the pure stereoisomers of formulae (Xa) and (Xb) using methods known to the person skilled in the art, such as fractionised crystallisation or column chromatography on silica gel.

Scheme 4

(lib) or (llb-ent) (lie) or (llc-ent) Scheme 4 illustrates the transformation of the enatiomerically pure stereoisomer (Xa) or (Xb) to compounds of formula (I la) or (lla-ent), in which R E represents a d-Ce-alkyl group, and in which Y stands for a leaving group LG or a hydroxy I group, and whereby (I la) and (ent-lla) refer to the two enantiomers of the structure shown. The enatiomerically pure stereoisomer (I la) or (lla-ent) can subsequently be further transformed into the compounds of the present invention as outlined in Scheme 1. Said transformation can be accomplished by various ways known to the person skilled in the art; preferably, intermediates of the formula (Xa) or (Xb) are subjected to a transesterification reaction using, for example,

titanium(IV)tetraethanolate in ethanol preferentially at elevated temperature. The resulting pyrimidine based ester synthons as pure stereoisomers of formula (I la) or (lla-ent) can subsequently be subjected to mild hydrolysis, as discussed supra, to give enantiopure carboxylic acids of formula (lib) or (llb-ent).

For example, further elaboration of compounds of formulae (lib) or (llb-ent), e.g. into compounds of the formulae (lie) or (llc-ent), in which R 3 stands for -C(=0)NR 5 R 4 , can be accomplished by coupling with amines of formula HN(R 5 )R 4 , in which R 4 and R 5 have the meaning as given for general formula (I) and which are widely commercially available, with a suitable coupling agent, such as HATU, TBTU, or 2,4,6-tripropyl-1 ,3,5,2,4.6- trioxatriphosphinane 2,4.6-trioxide (also known as T3P), as outlined in Scheme 4, to eventually give enantiopure amides of the general formula (I).

If needed, compounds of formulae (Ms), (lla-ent), (lla-rac), (lib), (llb-ent), (llb-rac), (lie), (llc- ent), (Vll-rac), (IX), (Xa) and (Xb), in which Y represents a hydroxy group can be converted into the respective compounds in which Y stands for a leaving group LG, i.e. into compounds of formulae (II) referred to in Schemes 1 and 2, by the methods described supra.

Scheme 5

Ar-S0 3 NH 2

(XVI) (III)

Compounds of the formula (III) are commercially available or can be synthesised by adapting procedures known to the person skilled in the art (see e.g. J. D. Kendall et al., Bioorganic & Medicinal Chemistry Vol. 20 (2012) 69-85; J. D. Kendall Current Organic Chemistry, 2011 , 15, 2481 -2518). Scheme 5 exemplifies one route that allows variations and modifications in 1 starting from commercially available 4-amino-pyridines of the formula (XI). The amino group in (XI) may be optionally protected by a suitable protective group (PG) like, for example, a tert- butyloxycarbonyl- or a allyloxycarbonyl-group, to give compounds of the formula (XII).

N-Amination to compounds of the formula (XIV) is achieved by reacting compounds of the formula (XII) with commercially available aminooxy-sulfonyl compounds of the formula (XIII) in which Ar represents a optionally substituted aryl group, like, for example, 2- [(aminooxy)sulfonyl]-1 ,3,5-trimethylbenzene. This transformation can also be achieved by reacting compounds of the formula (XII) with 0-(2,4-dinitrophenyl)hydroxylamine.

Compounds of the formula (XIV) react with propiolate compounds of formula (XV) in which R E represents a hydrogen or a d-Ce-alkyl group optionally in the presence of an organic or inorganic base to compounds of formula (XVI). Cleavage of the ester (R E = hydrogen), decarboxylation and removal of the protective group (PG) give compounds of formula (III). However, also other routes may be used to synthesise the target compounds (III), in accordance with common general knowledge of a person skilled in the art of organic synthesis. The order of transformations exemplified in Scheme 5 is therefore not intended to be limiting. In addition, interconversion of any of the substituents as defined herein for R\ PG and R E can be achieved before and/or after the exemplified transformations as described supra.

In the present text, in particular in the Experimental Section, for the synthesis of

intermediates and of examples of the present invention, when a compound is mentioned as a salt form with the corresponding base or acid, the exact stoichiometric composition of said salt form, as obtained by the respective preparation and/or purification process, is, in most cases, unknown.

Unless specified otherwise, suffixes to chemical names or structural formulae such as "hydrochloride", "trifluoroacetate", "sodium salt", or "x HCI", "x CF3COOH", "x Na * ", for example, are to be understood as not a stoichiometric specification, but solely as a salt form.

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. The lUPAC names of the examples and intermediates were generated using the program ' ACD/Name batch version 12.01 ' from ACD LABS, and were adapted if needed.

The various aspects of the invention described in this application are illustrated by the following examples which are not meant to limit the invention in any way.

Example 1

(7S)-4-[(4 hIoropyrazoSo[1,5-a]pyridin-5-yl)amino]-N,N-dimethyf-5,6,7,e - tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide

A mixture comprising 50 mg (125 μιηοΙ) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]- 5.6J,8-tetrahydro[1]benzothieno[2.3-d]pyrimidine-7-carboxyli c acid (prepared according to example 1 a), 2.44 ml. N,N-dimethylacetamide, 131 μΙ_ N-ethyl-N-isopropylpropan-2-amine. 312 μΙ_ N-methylmethanamine (2M in tetrahydrofuran) and 223 μΙ_ 2.4,6-tripropyl-1 ,3,5,2,4,6- trioxatriphosphinane 2.4,6-trioxide solution (50% in Ν,Ν-dimethylformamide) was stirred at 23°C for 16 hours. The crude mixture was concentrated, purified by chromatography and digestion with diethyl ether to give 36.0 mg (64%) of the title compound.

H-N R (400 MHz, DMSO-d6), δ [ppm]= 1.80 (1 H), 2.1 1 (1 H), 2.88 (3H), 2.91-3.04 (2H), 3.1 1 (3H), 3.21 (2H), 3.31 (1 H), 6.66 (1 H), 7.57 (1 H), 8.08 (1 H), 8.32 (1 H), 8.43 (1 H), 8.74

(1 H)

Example 1 a

(7S)-4-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8-tetrahydro[1]benzothr d]pyrimidine-7-carboxylic acid

A mixture comprising 392 mg (916 pmol) ethyl (7S)-4-[(4-chloropyrazolo[1.5-a]pyridin-5- yl)amino]-5.6,7,8-tetrahydro[1]benzothieno[2,3-d]pyrimidine- 7-carboxylate (prepared according to intermediate example 1 b) 16 mL tetrahydrofuran, 4 mL methanol and 5.5 ml_ lithium hydroxide solution (1 M in water) was stirred at 23°C for 1 hour. The mixture was acidified with hydrochloric acid and concentrated. The precipitate was filtered off, washed with water and dried to give 345 mg (80%) of the title compound.

Example 1 b

Ethyl (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylate

To a solution of 1.14 g (3.85 mmol) ethyl (7S)-4-chloro-5,6,7,8-tetrahydro[1]benzothieno[2,3- d]pyrimidine-7-carboxylate (prepared according to intermediate example 1 c) were added 43 mg palladium(ll) acetate, 180 mg 1 ,1 '-binaphthalene-2,2'-diylbis(diphenylphosphane), 646 mg 4-chloropyrazolo[1 ,5-a]pyridin-5-amine (prepared according to intermediate example 1f) and 1.88 g cesium carbonate. The mixture was stirred at 80°C for 16 h. Water was added, the mixture extracted with dichloromethane, the organic layer washed with hydrochloric acid (2M), water, brine and dried over sodium sulfate. After filtration and concentration the residue was crystallized from ethanol to give 395 mg (20%) of the title compound.

Example 1 c

-d]pyrimidine-7-carboxylate

A mixture comprising 27.6 g (64.6 mmol) (4S.5R)-3-{[(7S)-4-chloro-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl]carbonyl}-4-me thyl-5-phenyl-1 ,3-oxazolidin-2- one (prepared according to intermediate example 1d), 830 ml. ethanol and 24.4 ml_ titanium(4+) tetraethanolate was refluxed for 20 hours. 1.4 L ethyl acetate and 18 ml. water were added and the mixture was stirred for 30 minutes. Silica gel was added and stirring was continued for 10 minutes. The mixture was filtered through celite, the solvents were removed and the residue was purified by chromatography to give 18.8 g (93%) of the title compound.

Example 1d

(4S,5R)-3-{[(7S)-4-Chloro-5,6,7.8-tetrahydro[1]benzothien o[2,3-d]pyrimidin-7-yl]carbonyl}-4- methyl-5-phenyl-1 ,3-oxazolidin-2-one (A) and i4S.5R)-3-{[(7R)-4-chloro-5, 6,7,8- teirahydro[1]benzoihieno[2,3-d]pyrimidin-7-y!3carbonyl}-4-me ihyl-5-phenyl-1 ,3-oxazolidin-2- one (B)

To a solution of 26.8 g (4S,5 )-4-methyl-5-phenyl-1 ,3-oxazolidin-2-one in 428 mL tetrahydrofurane were added 70 mL n-buthyllithium (2.5 M in hexane) at -78°C and the mixture was stirred at -60°C for 1 hour. A solution of 45.8 g (159 mmol) 4-chloro-5.6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7(RS)-carbonyl chloride (prepared according to intermediate example 1 e) in 428 mL tetrahydrofurane was added and stirring was continued at -70°C for 1 hour. The mixture was poured into water, tetrahydrofurane was removed, the precipitate was filtered off, washed with water and resolved in dichloromethane. The organic layer was dried over sodium sulphate followed by addition of acetonitrile. The

dichloromethane was removed, the precipitate filtered, washed with acetonitrile and diethylether to give 27.6 g (38%) of the title compound A. From the mother liquor a second precipitate was obtained on standing overnight to give 25.5 g (35%) of the title compound B.

Example 1 e

-Chloro-5,6,7,8-tetrahydro[1]benzothieno[2,3-d]pyrimidine-7( RS)-carbonyl chloride

A mixture comprising 42.87 g (159 mmol) (RS)-4-chloro-5, 6,7,8- tetrahydro[1]benzothieno[2.3-d]pyrimidine-7-carboxylic acid (prepared according to

WO2013174744) and 349 mL thionyl chloride was heated at 100°C for 3 hours. The reagent was removed to give the title compound that was used without further purification.

Example 1f

4-Chloropyrazolo[1 ,5-a]pyridin-5-amine

A suspension of 5.0 g (20.9 mmol) ethyl 5-amino-4-chloropyrazolo[1 ,5-a]pyridine-3- carboxylate (prepared according to intermediate example 1g) in 50 mL sulfuric acid (40%) was stirred at 80°C for 4 hours. The solution was cooled to 10"C and then to 300 mL aqueous saturated potassium carbonate was added. The mixture was extracted with dichloromethane, the combined organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was diluted with concentrated hydrochloric acid, the solvent was removed at reduced pressure to afford the crude product which was purified by chromatography to give 2.35 g (67%) of the title compound.

Example 1g

Ethyl 5-amino-4-chloropyrazolo[1 ,5-a]pyridine-3-carboxylate

A mixture of 16 g (44 mmol) ethyl 5-(tert-butoxycarbonylamino)-6-chloro-pyrazolo[1.5- a]pyridine-3-carboxylate and ethyl 5-(tert-butoxycarbonylamino)-4-chloro-pyrazolo[1 ,5- a]pyridine-3-carboxylate (prepared according to intermediate example 1 h) in 50 ml_ dichloromethane and 50 mL trifluoroacetic acid was stirred at 10°C for 2 hous. The residue was diluted with aqueous saturated potassium carbonate, extracted with dichloromethane and concentrated. The crude product was purified by column chromatography to give 5.0 g (46%) of the title compound.

Example 1 h

Ethyl 5-(tert-butoxycarbonylamino)-6-chloro-pyrazolo[1 ,5-a]pyridine-3-carboxylate and ethyl 5-(tert-butoxycarbonylamino)-4-chloro-pyrazolo[1 ,5-a]pyridine-3-carboxylate

To a solution of 25 g crude 1 -amino-4-((tert-butoxycarbonyl)amino)-3-chloropyridin-1 -ium 2,4-dinitrophenolate (prepared according to intermediate example 1 i) in 100 mL N,N- dimethylformamide was added 24.2 g potassium carbonate. The mixture was stirred at 20°C for 1 h, 5.73 g ethyl propiolate was added and the mixture was stirred at 20°C for 18h. The mixture was diluted with water, extracted with ethyl acetate and concentrated. The residue was purified by chromatography to give 8.0 g (60%) of a mixture of the title compounds.

Example 1 i

1 -Amino-4-((tert-butoxycarbonyl)amino)-3-chloropyridin-1-ium 2,4-dinitrophenolate

A solution of 9.0 g (39.4 mmol) tert-butyl N-(3-chloro-4-pyridyl)carbamate (prepared according to WO2008130021 ) and 15.7 g 0-(2 ! 4-dinitrophenyl)hydroxylamine in 120 mL acetonitriie was stirred at 50°C for 40 h. The mixture was evaporated under reduced pressure to afford 25 g of the crude product which was used without further purification.

Example 2

(7S)-4-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl[3-{dimethylami no)azetidin-1- yl]methanone

50 mg (125 μιτιοΙ) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxyiic acid (prepared according to intermediate example 1 a) were transformed in analogy to example 1 using 3- (dimethylammonio)azetidinium dichioride to give after working up and purification 41.6 mg (66%) of the title compound.

1 H- MR (400 MHz, DMSO-d6), δ [ppm]= 1.80 (1 H), 2.10 (7H), 2.80 (1 H), 2.93 (2H), 3.05 (1 H), 3.16 (1 H), 3.31 (1 H), 3.67 (1 H), 3.89 (1 H), 4.05 (1 H), 4.26 (1 H), 6.66 (1 H), 7.58 (1 H), 8.08 (1 H), 8.32 (1 H), 8.43 (1 H), 8.74 (1 H).

Example 3

(7S)-4-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-N-{2-methoxyethyl)-N-methyl-

5,6,7,8-tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carbo xamide

50 mg (125 μιηοΙ) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 1 a) were transformed in analogy to example 1 using 2-methoxy-N- methylethanamine to give after working up and purification 24.2 mg (37%) of the title compound.

1 H- MR (400 MHz, DMSO-d6), δ [ppm]= 1.81 (1 H), 2.10 (1 H), 2.89+3.14 (3H), 2.91-3.04 (2H), 3.16-3.36 (3H), 3.27+3.28 (3H), 3.43-3.68 (4H), 6.66 (1 H), 7.57 (1 H), 8.08 (1 H), 8.31 (1 H), 8.43 (1 H), 8.74 (1 H)

Example 4

(7S)-4-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- ieirahydro[1]benzothieno[2,3-d]pyrimidin-7-y![(3R)-3-(dimeth yfamino)pyrroSidin-1 - yl]methanone

50 mg (125 pmol) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2.3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 1 a) were transformed in analogy to example 1 using (3R)-N,N- dimethylpyrrolidin-3-amine to give after working up and purification 34.2 mg (52%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.59-1.86 (2H), 2.00-2.21 (2H), 2.17+2.18 (6H), 2.66 (1 H), 2.93-3.68 (8H), 3.80+3.90 (1 H), 6.66 (1 H), 7.54+7.58 (1 H), 8.08 (1 H), 8.32 (1 H), 8.43 (1 H). 8.74 (1 H)

Example 5

(7S)-4-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl[(2R,6S)-2,6-di methylmorpholin-4- yl]methanone

50 mg (125 pmol) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 1 a) were transformed in analogy to example 1 using (2R,6S)-2,6- dimethylmorpholine to give after working up and purification 39.7 mg (61 %) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.08-1.15 (6H), 1.84 (1 H), 2.09 (1 H), 2.28 (1 H), 2.77 (1 H), 2.90-3.07 (2H), 3.17-3.33 (3H), 3.45 (1 H), 3.54 (1 H), 3.99 (1 H), 4.32 (1 H), 6.67 (1 H), 7.54 (1 H), 8.08 (1 H), 8.33 (1 H), 8.42 (1 H), 8.75 (1 H)

Example 6

(7S)-4-[(4-ChloropyrazoSo[1,5-a]pyridin-5-yf)amino]-5,6,7,8- ietrahydro[1]benzothieno[2,3-d]pyrimidin-7-yf4-[2-(dimethyfa mino)ethyl3piperazin-1 - ylmethanone

50 mg (125 μιτιοΙ) (7S)-4-[(4-chloropyrazolo[1 , 5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 1 a) were transformed in analogy to example 1 using N,N-dimethyl-2- (piperazin-l-yl)ethanamine to give after working up and purification 30.3 mg (43%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.82 (1 H), 2.08 (1 H), 2.14 (6H), 2.33-2.47 (7H), 2.53 (1 H), 2.88-3.05 (2H), 3.15-3.39 (3H), 3.50 (2H), 3.57 (2H), 6.65 (1 H), 7.56 (1 H), 8.08 (1 H), 8.31 (1 H), 8.42 (1 H), 8.74 (1 H)

Example 7

(7S)- -[(4- ethoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-N,N-dimethyl-5 > 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide

50 mg (126 μιηοΙ) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2.3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 to give after working up and purification 38.7 mg (69%) of the title compound. 1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.82 (1 H), 2.15 (1 H), 2.89 (3H), 2.91-3.02 (2H), 3.12 (3H), 3.16-3.32 (3H), 4.02 (3H), 6.72 (1 H), 7.87 (1 H), 7.98 (1 H), 8.20 (1 H), 8.45 (1 H), 8.53 (1 H)

Example 7a

(7S)-4-[(4-Methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6J,8-tetra ydro[1]benzothii djpyrimidine-7-carboxylic acid

563 mg (1.33 mmol) ethyl (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylate (prepared according to intermediate example 7b) were transformed in analogy to example 1 a to give after working up 558 mg (100%) of the title compound.

Example 7b

Ethyl (7S)-4-[(4-meihoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]

tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylate

1.05 g (3.55 mmol) ethyl (7S)-4-chloro-5 ! 6 ! 7.8-tetrahydro[1]benzothieno[2,3-d]pyrimidine-7- carboxylate (prepared according to intermediate example 1 c) were transformed in analogy to example 1 b using 4-methoxypyrazolo[1 ,5-a]pyridin-5-amine (prepared according to intermediate example 7c) to give after working up 566 mg (36%) of the title compound. Example 7c

4-Methoxypyrazolo[1 ,5-a]pyridin-5-amine

4.40 g (13.1 mmol) ethyl 5-[(tert-butoxycarbonyl)amino]-4-methoxypyrazolo[1.5-a]pyrid ine-3- carboxylate (prepared according to intermediate example 7d) were transformed in analogy to intermediate example 1f to give after working up 2.02 g (77%) of the title compound isolated as HCI salt.

Example 7d

Ethyl 5-[(tert-butoxycarbonyl)amino]-4-methoxypyrazolo[1 : 5-a]pyridine-3-carboxylate

28 g crude 1 -amino-4-[(tert-butoxycarbonyl)amino]-3-methoxypyridinium 2,4-dinitrophenolate (prepared according to intermediate example 7e) were transformed in analogy to intermediate example 1 h to give after working up 5.0 g of the title compound.

Example 7e

1-Amino-4-[(tert-butoxycarbonyl)amino]-3-methoxypyridinium 2.4-dinitrophenolate

10.0 g (44.6 mmol) tert-butyl (3-methoxypyridin-4-yl)carbamate (prepared according to intermediate example 7f) were transformed in analogy to intermediate example 1 i to give after working up 56 g crude title compound. Example 7f

tert-Butyl (3-methoxypyridin-4-yl)carbamate

To a suspension of 25.0 g 3-methoxypyridin-4-amine (201.39 mmol) in 100 mL

tetrahydrofurane was added 402. 8 mL lithium-bis(trimethylsilyl)amide (1 M in THF) at 0°C. After stirring for 1 h, 48.4 g di-tert-butyl dicarbonate was added to the reaction mixture. The reaction mixture was stirred at 0 C for 3 h, poured into saturated ammoniumchloride and extracted with ethyl acetate. The combined organic layer was dried over magnesium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography to give 37 g of crude product, which was triturated with petroleum ether to give 20 g of the title compound. Example 8

[3-(Dimethylamino)azetidin-1 -yl](7S)^-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]- 5,6,7, 8-tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-ylmethanone

50 mg (126 μηηοΙ) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5.6.7.8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using 3- (dimethylammonio)azetidinium dichloride to give after working up and purification 43.8 mg (69%) of the title compound.

1 H-N (400 MHz, DMSO-d6). δ [ppm]= 1.81 (1 H), 2.10 (6H), 2.13 (1 H), 2.80 (1 H), 2.87- 2.99 (2H), 3.06 (1 H), 3.17 (1 H), 3.29 (1 H), 3.67 (1 H), 3.90 (1 H), 4.02 (3H), 4.09 (1 H). 4.26 (1 H), 6.73 (1 H), 7.88 (1 H), 7.98 (1 H), 8.20 (1 H), 8.46 (1 H). 8.53 (1 H)

Example 9

(7S)-N-{2- ethoxyethyl)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-N-methyl- 5,6,7, 8-tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide

50 mg (126 μιτιοΙ) (7S)-4-[(4-methoxypyrazolo[1.5-a]pyridin-5-yl)amino]-5.6,7.8 - tetrahydro[1]benzothieno[2.3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using 2-methoxy-N- methylethanamine to give after working up and purification 37.4 mg (60%) of the title compound.

H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.84 (1 H), 2.13 (1 H), 2.89+3.14 (3H), 2.91-3.02 (2H), 3.16-3.31 (3H), 3.27+3.29 (3H), 3.42-3.71 (4H), 4.02+4.03 (3H), 6.72 (1 H), 7.86+7.89 (1 H), 7.98 (1 H), 8.20 (1 H), 8.45 (1 H), 8.53 (1 H) Example 10 [(3R)-3^Dimethylamino)pyrrolidin-1-yl](7S)-4-[(4-methoxypyra zolo[1 ,5-a]pyridin-5- yl)amino]-5,6J,8-tetrahydro[1]benzothieno[2,3-d]pyrimidin-7- ylmethanone

50 mg (126 pmol) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5.6J,8- tetrahydro[1]benzothieno[2.3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using (3R)-N,N- dimethylpyrrolidin-3-amine to give after working up and purification 35.0 mg (53%) of the title compound.

1H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.60-1.88 (2H), 2.00-2.16 (2Η), 2.17+2.18 (6H), 2.62+2.70 (1 H), 2.93-3.67 (8H), 3.79+3.90 (1 H), 4.02 (3H), 6.72 (1 H), 7.84+7.87 (1 H), 7.98 (1 H), 8.20 (1 H), 8.45 (1 H), 8.53 (1 H)

Example 11

[(2R,6S)-2,6-Dimethylmorpholin-4-yl](7S)-4-[(4-methoxypyr azolo[1 ,5-a]pyridin-5- yl)amino]-5,6,7,8-tetrahydro[1]benzothieno[2,3-d]pyrimidin-7 -ylmethanone

50 mg (126 pmol) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using (2R,6S)-2,6- dimethylmorpholine to give after working up and purification 26.7 mg (41 %) of the title compound.

H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.12 (6H), 1.86 (1 H), 2.1 1 (1 H), 2.29 (1 H), 2.78 (1 H), 2.90-3.06 (2H), 3.17-3.32 (3H), 3.45 (1 H), 3.54 (1 H), 3.98 (1 H), 4.02 (3H), 4.33 (1 H), 6.73 (1 H), 7.83 (1 H), 7.99 (1 H), 8.19 (1 H), 8.45 (1 H), 8.53 (1 H)

Example 12

4-[2-{Dimethylamino)ethyl]piperazin-1-yl(7S)-4-[(4-methoxypy razolo[1 ,5-a]pyridin-5- yl)amino]-5,6,7,8-tetrahydro[1]benzothieno[2,3-d]pyrimidin-7 -ylmethanone

50 mg (126 μιηοΙ) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6J,8- tetrahydro[1]benzothieno[2.3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using N,N-dimethyl-2- (piperazin-l-yl)ethanamine to give after working up and purification 51.0 mg (72%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.85 (1 H), 2.1 1 (1 H), 2.14 (6H), 2.26-2.49 (8H), 2.88-3.04 (2H), 3.17-3.30 (3H), 3.50 (2H), 3.58 (2H), 4.02 (3H), 6.73 (1 H), 7.86 (1 H), 7.99 (1 H), 8.19 (1 H), 8.45 (1 H), 8.53 (1 H)

Example 13

(4-{[2^Dimethylamino)ethyl](methyl)amino}piperidin-1-yl){(7S )- -[(4- methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8-tetrahydro[1]benzoth

d]pyrimidin-7-yl}methanone

50 mg (126 pmol) (7S)-4-[(4-methoxypyrazolo[1.5-a]pyridin-5-yl)amino]-5.6.7.8 - tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using 4-{[2- (dimethylammonio)ethyl](methyl)ammonio}piperidinium trichloride to give after working up and purification 39 mg (52%) of the title compound.

1 H-NMR (400 MHz. DMSO-d6), δ [ppm]= 1.24 (1 H), 1.39 (1 H), 1.69-1.80 (2H), 1.84 (1 H), 2.10 (1 H) 2.13 (6H). 2.18 (3H). 2.28 (2H), 2.45-2.62 (4H), 2.88-3.10 (3H), 3.18-3.33 (3H), 4.02 (3H), 4.08 (1 H), 4.48 (1 H), 6.72 (1 H), 7.85 (1 H), 7.98 (1 H), 8.18 (1 H), 8.44 (1 H), 8.52

(1 H)

Example 14

{(7S)-4-[(4-Methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}(4-methylpiper azin-1 -yl)methanone

50 mg (126 μιτιοΙ) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using 1- methylpiperazine to give after working up and purification 54 mg (85%) of the title compound. 1 H-N R (400 MHz, DMSO-d6), δ [ppm]= 1.84 (1 H), 2.07-2.16 (1 H), 2.20 (3H), 2.28 (2H), 2.35 (2H), 2.89-3.02 (2H), 3.16-3.30 (3H), 3.51 (2H), 3.58 (2H), 4.02 (3H), 6.72 (1 H), 7.86 (1 H). 7.98 (1 H), 8.18 (1 H). 8.45 (1 H), 8.52 (1 H) Example 15

(3-Hydroxy-3-methylazetidin-1-yl){(7S)-4-[(4-methoxypyrazolo [1 ,5-a]pyridin-5- -yl}methanone

50 mg (126 pmol) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6.7.8- tetrahydro[1]benzothieno[2.3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using 3-hydroxy-3- methylazetidinium chloride to give after working up and purification 46 mg (74%) of the title compound.

1 H- MR (400 MHz, DMSO-d6), δ [ppm]= 1.40 (3H), 1.81 (1 H), 2.13 (1 H), 2.79 (1 H), 2.87- 2.97 (2H). 3.18 (1 H), 3.28 (1 H), 3.70-3.77 (2H). 4.02 (3H). 4.03-4.13 (2H), 5.68 (1 H), 6.72 (1 H), 7.86 (1 H), 7.98 (1 H), 8.18 (1 H), 8.45 (1 H), 8.52 (1 H)

Example 16

(7S)-N-[2-{Dimethylamino)ethyl]-4-[(4-methoxypyrazolo[1 ) 5-a]pyridin-5-yl)amino]- -tetrahydro[1]benzothieno -d]pyrimidine-7-carboxamide

50 mg (126 μιτιοΙ) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5.6.7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using N,N- dimethylethane-1.2-diamine to give after working up and purification 49 mg (79%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.86 (1 H), 2.15 (7H), 2.30 (2H), 2.70 (1 H), 2.92- 2.98 (2H), 3.1 1 -3.30 (4H), 4.02 (3H), 6.71 (1 H), 7.90 (1 H), 7.96 (1 H), 7.97 (1 H). 8.19 (1 H), 8.45 (1 H), 8.52 (1 H) Example 17

{(7S)- -[(4-Methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(1S,4S)-5-met hyl-2,5-

50 mg (126 pmol) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7.8- tetrahydro[1]benzothieno[2.3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using (1 S.4S)-2-methyl- 2,5-diazoniabicyclo[2.2.1]heptane dibromide to give after working up and purification 21 mg (34%) of the title compound.

1H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.65 (1 H), 1.76-1.93 (2H), 2.1 1 (1 H), 2.31 +2.32

(3H), 2.73-3.70 (10H), 4.01 (3H), 4.53+4.60 (1 H), 6.72 (1 H), 7.83+7.88 (1 H), 7.98 (1 H), 8.19 (1 H), 8.45 (1 H), 8.52 (1 H)

Example 18

{(7S)-4-[(4-Methoxypyrazolo[1,5-a]pyridin-5-yl)amino]-5,6 ,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(1 R,4R)-5-methyl-2,5-

50 mg (126 pmol) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using (1 R,4R)-2-methyl- 2,5-diazoniabicyclo[2.2.1]heptane dichloride to give after working up and purification 25 mg (41 %) of the title compound.

1 H- MR (400 MHz, DMSO-d6). δ [ppm]= 1.60+1.70 (1 H). 1.75-1.89 (2H), 2.13 (1 H), 2.30+2.34 (3H), 2.73-3.05 (4H), 3.10-3.66 (6H), 4.03 (3H), 4.54+4.59 (1 H), 6.72 (1 H), 7.88+7.90 (1 H), 7.98 (1 H), 8.19 (1 H), 8.45 (1 H), 8.52 (1 H)

Example 19

{(7S)-4-[(4-Methoxypyrazofo[1 ,5-a]pyridin-5-yf)amino]-5,6,7,B- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(1S,4S)-2-oxa -5-azabicyclo[2.2.1]hept-5-

50 mg (126 pmol) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6J,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using (1 S.4S)-2-oxa-5- azoniabicyclo[2.2.1 Jheptane chloride to give after working up and purification 31 mg (52%) of the title compound.

H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.77-1.94 (3H), 2.13 (1 H), 2.82-3.32 (6H),

3.54+3.66 (1 H), 3.59+3.73 (1 H), 3.77 (1 H), 4.01 (3H), 4.62+4.67 (1 H), 4.77+4.88 (1 H), 6.72 (1 H), 7.82+7.88 (1 H), 7.98 (1 H), 8.19 (1 H), 8.45 (1 H), 8.52 (1 H)

Example 20

{(7S)-4-[(4- ethoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(1 R,4R)-2-oxa-5-azabicyclo[2.2.1]hept- -yl]methanone

50 mg (126 μιτιοΙ) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using (1 R,4R)-2-oxa-5- azoniabicyclo[2.2.1 ]heptane chloride to give after working up and purification 32 mg (52%) of the title compound. 1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.77-1.91 (3H), 2.17 (1 H), 2.78-3.32 (6H), 3.52+3.65 (1 H), 3.60+3.77 (1 H), 3.74 (1 H), 4.03 (3H), 4.62+4.68 (1 H), 4.78+4.86 (1 H), 6.72 (1 H), 7.89+7.91 (1 H), 7.98 (1 H), 8.20 (1 H), 8.45 (1 H), 8.52 (1 H)

Example 21

[4-(Dimethylamino)piperidin-1-yl]{(7S)^-[(4-methoxypyrazolo[ 1 ,5-a]pyridin-5-yl) -tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}methanone

50 mg (126 μητιοΙ) (7S)-4-[(4-methoxypyrazolo[1.5-a]pyridin-5-yl)amino]-5.6J.8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using N,N- dimethylpiperidin-4-amine to give after working up and purification 39 mg (60%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.21 (1 H), 1.35 (1 H), 1.74-1.89 (3H), 2.1 1 (1 H), 2.18 (6H), 2.33 (1 H), 2.62 (1 H), 2.88-3.13 (3H), 3.18-3.31 (3H), 4.02 (3H), 4.06 (1 H), 4.41 (1 H). 6.72 (1 H), 7.85 (1 H). 7.98 (1 H), 8.18 (1 H), 8.44 (1 H), 8.52 (1 H)

Example 22

{(7S)-4-[(4-Methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(3S)-3-methyl morpholin-4-

50 mg (126 μιτιοΙ) (7S)-4-[(4-methoxypyrazolo[1.S-aJpyridin-S-y aminoj-S^J.e- tetrahydro[1]benzothieno[2.3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using (3S)-3- methylmorpholine to give after working up and purification 37 mg (62%) of the title compound.

H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.16+1.30 (3H), 1.82 (1 H), 2.05-2.18 (1 H), 2.85- 4.48 (12H). 4.02 (3H), 6.72 (1 H), 7.84-7.91 (1 H), 7.98 (1 H). 8.18 (1 H), 8.45 (1 H). 8.52 (1 H) Example 23

{(7S)-4-[(4- ethoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(3R)-3-methyl morpholin-4-

50 mg (126 pmol) (7S)-4-[(4-met oxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5.6,7.8- tetra ydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using (3R)-3- methylmorpholine to give after working up and purification 19 mg (31 %) of the title compound.

H- MR (400 MHz, DMSO-d6), δ [ppm]= 1.16+1.32 (3H), 1.90 (1 H), 2.10 (1 H), 2.87-4.46 (12H), 4.02 (3H), 6.72 (1 H), 7.81-7.88 (1 H), 7.98 (1 H), 8.19 (1 H), 8.45 (1 H), 8.52 (1 H)

Example 24

{(7S)-4-[(4- ethoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- -yl)methanone

50 mg (126 μιηοΙ) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using morpholine to give after working up and purification 24 mg (40%) of the title compound.

1H-NMR (400 MHz. DMSO-d6), δ [ppm]= 1.85 (1 H), 2.13 (1 H). 2.90-3.04 (2H). 3.17-3.32 (3H), 3.51 (2H), 3.54-3.64 (6H), 4.02 (3H), 6.72 (1 H). 7.85 (1 H). 7.98 (1 H), 8.18 (1 H), 8.45 (1 H), 8.52 (1 H)

Example 25

[(3S)-3-{Dimethylamino)pyrrolidin-1 -yl]{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5- yl)amino]-5,6,7,8-tetrahydro[1]benzothieno[2,3-d]pyrimidin-7 -yl}methanone

50 mg (126 pmol) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6.7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using (3S)-N,N- dimethylpyrrolidin-3-amine to give after working up and purification 32 mg (52%) of the title compound.

1 H-N R (400 MHz ,DMSO-d6). δ [ppm]= 1.58-1.87 (2H), 1.99-2.16 (2H), 2.17 (3H), 2.18 (3H), 2.61 +2.73 (1 H), 2.91-3.67 (8H), 3.75+3.83 (1 H), 4.02 (3H), 6.72 (1 H), 7.85+7.89 (1 H). 7.98 (1 H), 8.19 (1 H), 8.45 (1 H), 8.52 (1 H)

Example 26

1-{{(7S)- -[(4-Methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- idine-3-carbonitrile

50 mg (126 pmol) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6J,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using 3- cyanoazetidinium chloride to give after working up and purification 28 mg (49%) of the title compound.

H-NMR (400 MHz ,DMSO-d6), δ [ppm]= 1.80 (1 H), 2.14 (1 H), 2.78 (1 H), 2.85-3.03 (2H), 3.16 (1 H), 3.24-3.31 (1 H), 3.82 (1 H), 4.01 +4.02 (3H), 4.04+4.07 (1 H), 4.19 (1 H), 4.46-4.60 (2H), 6.72 (1 H), 7.85 (1 H), 7.98 (1 H), 8.19 (1 H), 8.45 (1 H), 8.52 (1 H)

Example 27

(7S)-N-[3-{Dimethylamino)propyl]-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-N- arboxamide

50 mg (126 μητιοΙ) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5.6.7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using Ν,Ν,Ν'- trimethylpropane-1 ,3-diamine to give after working up and purification 28 mg (44%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.60+1.69 (2H), 1.76-1.95 (1 H), 2.07-2.22 (3H), 2.1 1 +2.12 (6H), 2.86+3.10 (3H), 2.89-3.00 (2H), 3.12-3.48 (5H), 4.01 +4.02 (3H), 6.72 (1 H). 7.86+7.88 (1 H). 7.98 (1 H), 8.19 (1 H), 8.45 (1 H), 8.52 (1 H) Example 28

(7S)-N-[2^Dimethylamino)ethyl]-N-ethyl- -[(4-methoxypyrazolo[1 ,5-a]pyridin-5- -carboxamide

50 mg (126 μιτιοΙ) (7S)-4-[(4-methoxypyrazolo[1 , 5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using N'-ethyl-N,N- dimethylethane-1 ,2-diamine to give after working up and purification 20 mg (31 %) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.05+1.16 (3H), 1.86 (1 H), 2.10 (1 H), 2.17+2.18 (6H), 2.34 (1 H), 2.42 (1 H), 2.88-3.04 (2H), 3.08 (1 H), 3.19-3.51 (6H), 4.02 (3H), 6.72 (1 H), 7.88 (1 H), 7.98 (1 H), 8.19 (1 H), 8.45 (1 H), 8.52 (1 H)

Example 29

(7S)-N-[2-{Dimethylamino)ethyl]-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-N- arboxamide

50 mg (126 pmol) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7.8- tetrahydro[1]benzothieno[2.3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using Ν,Ν,Ν'- trimethylethane-1 ,2-diamine to give after working up and purification 31 mg (51 %) of the title compound. 1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.84 (1 H), 2.13 (1 H), 2.20+2.21 (6H), 2.38-2.46 (2H), 2.88+3.1 1 (3H), 2.90-3.03 (2H), 3.13-3.53 (5H), 4.01 +4.02 (3H), 6.72 (1 H), 7.86+7.88 (1 H), 7.98 (1 H), 8.19 (1 H), 8.45 (1 H), 8.52 (1 H) Example 30

(7S)-N^2-Hydroxyethyl)^-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-N-methyl- -tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide

50 mg (126 μιτιοΙ) (7S)-4-[(4-methoxypyrazolo[1 , 5-a]pyridin-5-yl)amino]-5, 6,7,8- tetra ydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using 2-

(methylamino)ethanol to give after working up and purification 17 mg (29%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.84 (1 H), 2.15 (1 H), 2.86+3.15 (3H), 2.91-3.01

(2H), 3.16-3.59 (7H), 4.01 +4.02 (3H), 4.68+4.87 (1 H), 6.72 (1 H), 7.85+7.88 (1 H), 7.98 (1 H), 8.19 (1 H), 8.45 (1 H), 8.52 (1 H)

Example 31

(7S)-N-(2,2-Diffuoroethyf)-4-[(4-methoxypyrazoSo[1,5-a]pyrid in-5-yS)amino]-N-methyS- -tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide

50 mg (126 μητιοΙ) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using 2,2-difluoro-N- methylethanamine to give after working up and purification 19 mg (32%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.85 (1 H), 2.1 1 +2.17 (1 H), 2.88-3.03 (2H), 2.96+3.21 (3H), 3.17-3.31 (3H), 3.64-4.00 (2H), 4.01 +4.02 (3H), 6.14+6.16 (1 H), 6.72 (1 H), 7.84+7.88 (1 H), 7.98 (1 H), 8.19 (1 H), 8.45 (1 H), 8.52 (1 H)

Example 32 (7S)-N-(2-Methoxyethyl)-4-[(4-methoxypyrazolo[1,5-a]pyridin- 5-yl)amino]-5,6,7,8-

50 mg (126 μητιοΙ) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8- tetra ydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using 2- methoxyethanamine to give after working up and purification 33 mg (58%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.87 (1H), 2.16 (1H), 2.72 (1H), 2.95 (2H), 3.14 (1H), 3.19-3.31 (3H), 3.27 (3H), 3.38 (2H), 4.02 (3H), 6.71 (1H), 7.89 (1H), 7.98 (1H), 8.12 (1H), 8.20 (1H), 8.45 (1H), 8.52 (1H)

Example 33

(7S)-N-lsopropyl- -[(4-methoxypyrazolo[1 ) 5-a]pyridin-5-yl)amino]-5 ) 6,7,8-

50 mg (126 pmol) (7S)-4-[(4-methoxypyrazolo[1 ^-alpyridin-S-y aminoj-S.e ^- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using propan-2-amine to give after working up and purification 31 mg (56%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.09 (6H), 1.86 (1H), 2.15 (1H), 2.62 (1H), 2.95 (2H), 3.13 (1H), 3.26 (1H), 3.88 (1H), 4.02 (3H), 6.72 (1H), 7.87 (1H), 7.91 (1H), 7.98 (1H), 8.20 (1H), 8.46 (1H), 8.52 (1H) Example 34

(7S)-4-[(4-Methoxypyrazolo[1,5-a]pyridin-5-yl)amino]-N-methy l-5,6,7,8- e

50 mg (126 μιτιοΙ) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5.6.7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using methanamine to give after working up and purification 26 mg (50%) of the title compound.

'H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.87 (1 H), 2.16 (1 H), 2.63 (3H), 2.67 (1 H), 2.96 (2H), 3.14 (1 H), 3.27 (1 H), 4.01 (3H), 6.71 (1 H), 7.87 (1 H), 7.95-8.00 (2H), 8.20 (1 H), 8.45 (1 H), 8.52 (1 H).

Example 35

(7S)-4-[(4-ChloropyrazoSo[1 ,5-a]pyridin-5-yl)amino]-N-isopropyl-5,6,7,8- e

50 mg (125 pmol) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a). were transformed in analogy to example 1 using propan-2-amine to give after working up and purification 42 mg (72%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.08 (6H), 1.84 (1 H), 2.1 1 (1 H), 2.63 (1 H), 2.95 (2H), 3.12 (1 H), 3.29 (1 H), 3.88 (1 H), 6.65 (1 H), 7.62 (1 H), 7.86 (1 H), 8.07 (1 H), 8.30 (1 H), 8.43 (1 H), 8.74 (1 H)

Example 36

(7S)-4-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-N-ethyl-N-methyl-5,6,7,8- de

50 mg (125 μιτιοΙ) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7.8- tetrahydro[1 ]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using N-methylethanamine to give after working up and purification 34 mg (59%) of the title compound.

H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.03+1.15 (3H), 1.75-1.91 (1 H), 2.08 (1 H), 2.85+3.07 (3H), 2.88-3.51 (7H), 6.65 (1 H), 7.57 (1 H), 8.07 (1 H), 8.30 (1 H), 8.42 (1 H), 8.74 (1 H) Example 37

(7S)^-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-N-(2-hydroxyethyl)-N-methyl- -tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide

50 mg (125 μηηοΙ) (7S)-4-[(4-c loropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7.8- tetra ydro[1]benzot ieno[2.3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using 2-(methylamino)ethanol to give after working up and purification 35 mg (57%) of the title compound.

1 H-NMR (400 MHz. DMSO-d6). δ [ppm]= 1.74-1.89 (1 H). 2.1 1 (1 H), 2.88+3.14 (3H), 2.90- 3.03 (2H), 3.15-3.58 (7H), 4.68+4.86 (1 H), 6.65 (1 H), 7.57 (1 H), 8.07 (1 H), 8.30 (1 H), 8.42 (1 H), 8.73 (1 H)

Example 38

(7S)-4-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-N-[2-{dimethylamino)ethyl]-N-ethyl- -tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxamide

50 mg (125 pmol) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using N'-ethyl-N,N-dimethylethane-1 ,2- diamine to give after working up and purification 42 mg (64%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.05+1.15 (3H), 1.84 (1 H), 2.07 (1 H), 2.17+2.18 (6H), 2.31 -2.45 (2H), 2.87-3.50 (9H), 6.65 (1 H), 7.58 (1 H), 8.07 (1 H), 8.30 (1 H), 8.43 (1 H), 8.74 (1 H) Example 39

1-({(7S)-4-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- zetidine-3-carbonitrile

50 mg (125 μητιοΙ) (7S)-4-[(4-chloropyrazolo[1 , 5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using 3-cyanoazetidinium chloride to give after working up and purification 36 mg (56%) of the title compound.

1H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.71-1.85 (1 H), 2.1 1 (1 H), 2.71 -3.04 (2H), 3.16 (1 H), 3.29 (1 H), 3.50 (1 H), 3.82 (1 H), 4.05 (1 H), 4.19 (1 H), 4.46-4.59 (2H), 6.66 (1 H), 7.56 (1 H), 8.08 (1 H), 8.32 (1 H), 8.42 (1 H), 8.74 (1 H)

Example 40

{(7S)-4-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}(3-hydroxy-3-m ethylazetidin-1 -

50 mg (125 pmol) (7S)-4-[(4-chloropyrazolo[1.5-a]pyridin-5-yl)amino]-5, 6,7.8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using 3-hydroxy-3-methylazetidinium chloride to give after working up and purification 24 mg (38%) of the title compound.

Ή-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.39 (3H), 1.80 (1 H), 2.10 (1 H), 2.79 (1 H), 2.93 (2H), 3.16 (1 H), 3.29 (1 H), 3.68-3.78 (2H), 4.01 -4.14 (2H), 5.68 (1 H), 6.66 (1 H), 7.54-7.60 (1 H), 8.08 (1 H), 8.31 (1 H), 8.43 (1 H), 8.74 (1 H)

Example 41

{(7S)-4-[(4-Chloropyrazolo[1,5-a]pyridin-5-yl)amino]-5,6,7,B - tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[4-{dimethylam ino)piperidin-1 -

50 mg (125 μιτιοΙ) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using N,N-dimethylpiperidin-4-amine to give after working up and purification 37 mg (55%) of the title compound. 1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.20 (1 H), 1.34 (1 H), 1.72-1.86 (3H), 2.06 (1 H), 2.17 (6H), 2.32 (1 H), 2.61 (1 H), 2.84-3.32 (6H), 4.05 (1 H), 4.41 (1 H), 6.59 (1 H), 7.54 (1 H), 8.03 (1 H), 8.34 (2H), 8.67 (1 H) Example 42

{(7S)-4-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}(4-{[2-

(

50 mg (125 μηηοΙ) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using 4-{[2-(dimethylammonio)ethyl](methyl)- ammoniojpiperidinium trichloride to give after working up and purification 47 mg (62%) of the title compound.

H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.24 (1 H), 1.39 (1 H), 1.67-1.87 (3H), 2.08 (1 H), 2.14 (6H), 2.18 (3H), 2.29 (2H), 2.45-2.62 (3H), 2.86-3.10 (4H), 3.16-3.30 (3H), 4.07 (1 H), 4.48 (1 H), 6.65 (1 H), 7.55 (1 H), 8.07 (1 H), 8.30 (1 H), 8.41 (1 H), 8.73 (1 H)

Example 43

{(7S)-4-[(4-Chloropyrazolo[1,5-a]pyridin-5-yl)amino]-5,6, 7,8- piperazin-1 -yl)methanone

50 mg (125 μιτιοΙ) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using 1-methylpiperazine to give after working up and purification 50 mg (79%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.82 (1 H), 2.08 (1 H), 2.20 (3H), 2.27 (2H), 2.34 (2H), 2.88-3.05 (2H), 3.14-3.31 (3H), 3.50 (2H), 3.58 (2H), 6.66 (1 H), 7.56 (1 H), 8.08 (1 H), 8.30 (1 H), 8.42 (1 H), 8.74 (1 H) Example 44

{(7S)-4-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(1S,4S)-2-oxa -5-azabicyclo[2.2.1]hept-5-

50 mg (125 pmol) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzoihieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using (1 S,4S)-2-oxa-5-azoniabicyclo[2.2.1]- heptane chloride to give after working up and purification 54 mg (84%) of the title compound. 1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.75-1.91 (3H), 2.10 (1 H), 2.81 -3.02 (3H), 3.09- 3.31 (3H), 3.54+3.66 (1 H), 3.58+3.73 (1 H), 3.76 (1 H), 4.61 +4.76 (1 H), 4.66+4.88 (1 H), 6.65 (1 H), 7.53+7.59 (1 H), 8.07 (1 H), 8.31 (1 H), 8.42 (1 H), 8.73 (1 H)

Example 45

{(7S)-4-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(1 R,4R)-2-oxa-5-azabicyclo[2.2.1]hept- -yl]methanone

50 mg (125 pmol) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using (1 R,4R)-2-oxa-5-azoniabicyclo[2.2.1]- heptane chloride to give after working up and purification 50 mg (79%) of the title compound. 1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.76-1.91 (3H), 2.13 (1 H), 2.79-3.32 (5H),

3.50+3.65 (1 H), 3.60+3.63 (1 H), 3.65+3.77 (1 H), 3.73 (1 H), 4.61 +4.78 (1 H), 4.67+4.86 (1 H), 6.65 (1 H), 7.60 (1 H), 8.07 (1 H), 8.30 (1 H), 8.43 (1 H), 8.74 (1 H)

Example 46

{(7S)-4-[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(1S,4S)-5-met hyl-2,5- diazabicyclo[2.2.1]hept-2-yl]methanone

50 mg (125 μιτιοΙ) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetra ydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using (1 S,4S)-2-methyl-2,5-diazoniabicyclo- [2.2.1]heptane dibromide to give after working up and purification 51 mg (79%) of the title compound.

1 H-N R (400 MHz, DMSO-d6), δ [ppm]= 1.64 (1 H), 1.75-1.92 (2H), 2.08 (1 H), 2.31 +2.32 (3H), 2.44+2.53 (1 H), 2.72-3.46+3.67 (9H), 4.52+4.60 (1 H), 6.65 (1 H), 7.53+7.59 (1 H), 8.07 (1 H), 8.30 (1 H), 8.42 (1 H), 8.73 (1 H)

Example 47

{(7S)- -[(4-Chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(1 R,4R)-5-methyl-2,5-

50 mg (125 pmol) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using (1 R,4R)-2-methyl-2,5-diazoniabicyclo- [2.2.1]heptane dichloride to give after working up and purification 36 mg (56%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.58-1.87 (3H), 2.09 (1 H), 2.30+2.33 (3H), 2.42+2.53 (1 H), 2.73-3.47+3.63 (9H), 4.54+4.58 (1 H), 6.63 (1 H), 7.59 (1 H), 8.06 (1 H), 8.30 (1 H), 8.40 (1 H), 8.71 (1 H) Example 48

{(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}(4-{[2-

(dimethylamino)ethyl](methyl)amino}piperidin-1-yl)methano ne

50 mg (125 μιηοΙ) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using N,N,N'-trimethylpropane-1 ,3-diamine to give after working up and purification 36 mg (56%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.60+1.68 (2H), 1.82 (1 H), 2.07 (1 H), 2.1 1 +2.13 (6H), 2.16-2.24 (2H), 2.85+3.09 (3H), 2.89-3.02 (2H), 3.12-3.48 (5H), 6.65 (1 H), 7.57 (1 H), 8.07 (1 H), 8.31 (1 H), 8.42 (1 H), 8.73 (1 H) Example 49

{(7S)- -[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3 l]pyrimidin-7-yl}[(3S)-3-(dimethylamino)pyrrolidin-1 -

50 mg (125 μιτιοΙ) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to example 1 a), were transformed in analogy to example 1 using (3S)-N,N-dimethylpyrrolidin-3-amine to give after working up and purification 37 mg (56%) of the title compound.

H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.58-1.85 (2H), 1.99-2.15 (3H), 2.18 (6H), 2.62+2.73 (1 H), 2.93-3.05 (3H), 3.15-3.68 (4H), 3.74+3.82 (1 H), 6.66 (1 H), 7.56+7.59 (1 H), 8.08 (1 H), 8.31 +8.32 (1 H), 8.42+8.43 (1 H), 8.74 (1 H)

Example 50

tert-Butyl [1 -{{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- ]carbamate

500 mg (1.26 mmol) (7S)-4-[(4-methoxypyrazolo[1 , 5-a]pyridin-5-yl)amino]-5, 6,7,8- tetra ydro[1 ]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using tert-butyl azetidin- 3-ylcarbamate hydrochloride to give after working up and purification 588 mg (80%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.39 (9H), 1.79 (1 H), 2.12 (1 H), 2.76 (1 H), 2.91 (2H), 3.17 (1 H), 3.28 (1 H), 3.73 (1 H), 4.01 (3H), 4.03 (1 H), 4.09 (1 H), 4.31 (1 H), 4.47 (1 H), 6.72 (1 H), 7.60 (1 H), 7.85 (1 H), 7.98 (1 H), 8.19 (1 H), 8.44 (1 H), 8.52 (1 H). Example 51

(3-Aminoazetidin-1 -yl){(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8-

632 mg (1.15 mmol) tert-butyl [1 -({(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]- 5,6J.8-tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}carbony l)azetidin-3-yl]carbamate

(prepared according to example 50) were transformed in analogy to intermediate example 1g at RT overnight and at 50°C for 3 hours to give after working up and purification 459 mg (84%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.80 (1 H), 2.05-2.31 (3H), 2.77 (1 H), 2.91 (2H), 3.17 (1 H), 3.28 (1 H), 3.50 (1 H), 3.70 (1 H), 3.83 (1 H), 4.01 (3H), 4.03 (1 H), 4.38 (1 H), 6.72 (1 H), 7.86 (1 H), 7.98 (1 H), 8.18 (1 H), 8.45 (1 H), 8.52 (1 H).

Example 52

(3-Aminoazetidin-1 -yl){(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8- hloride

To a solution of 100 mg (222 μιτιοΙ) (3-aminoazetidin-1 -yl){(7S)-4-[(4-methoxypyrazolo[1.5- a]pyridin-5-yl)amino]-5,6,7,8-tetrahydro[1]benzothieno[2,3-d ]pyrimidin-7-yl}methanone (prepared according to example 51 ) in 5 mL 1 ,4-dioxane were added 334 μΙ_ hydrogen chloride (1 M in 1 ,4-dioxane). The mixture was stirred at RT for 10 minutes and the solvents removed to give 1 16 mg (95%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6). δ [ppm]= 1.83 (1 H), 2.09-2.28 (1 H), 2.72-3.52 (5H), 3.80- 3.95 (1 H), 4.01 (3H), 4.04 (1 H), 4.13 (1 H), 4.23 (1 H), 4.53 (1 H), 6.72 (1 H), 7.82-7.91 (1 H), 7.98 (1 H), 8.15-8.24 (1 H), 8.31 -8.60 (5H)

Example 53

((7S)-4-[(4- ethoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-N- (pentacyclo[4.2.0.02,5.03,8.04,7]oct-1-yl)-5,6,7,8-tetrahydr o[1]benzothieno[2,3-

100 mg (253 μιτιοΙ) (7S)-4-[(4-methoxypyrazolo[1 , 5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1 ]benzothieno[2.3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using cuban-1-amine hydrochloride to give after working up and purification 94 mg (71 %) of the title compound. 1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.87 (1 H), 2.20 (1 H), 2.72 (1 H), 2.90-3.03 (2H), 3.15 (1 H), 3.26 (1 H), 3.85-3.90 (3H), 3.90-3.95 (1 H), 4.01 (3H), 4.04-4.10 (3H), 6.71 (1 H), 7.88 (1 H), 7.98 (1 H), 8.20 (1 H), 8.45 (1 H), 8.52 (1 H), 8.70 (1 H) Example 54

tert-Butyl [1 -({(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- l]carbamate

100 mg (250 μιηοΙ) (7S)-4-[(4-chloropyrazolo[1 , 5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1 ]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 1 a) were transformed in analogy to example 1 using tert-butyl azetidi 3-ylcarbamate hydrochloride to give after working up and purification 1 18 mg (81 %) of the title compound. 1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.39 (9H), 1.8 (1 H), 2.09 (1 H), 2.77 (1 H), 2.92 (2H), 3.17 (1 H), 3.28 (1 H), 3.73 (1 H), 3.98-4.13 (2H), 4.30 (1 H), 4.45 (1 H), 6.65 (1 H), 7.52-7.64 (2H), 8.07 (1 H), 8.31 (1 H), 8.42 (1 H), 8.73 (1 H) Example 55

(3-Aminoazetidin-1 -yl){(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8-

88.7 mg (160 pmol) tert-butyl [1 -({(7S)-4-[(4-chloropyrazolo[1.5-a]pyridin-5-yl)amino]-5.6.7 .8- tetrahydro[1]benzot ieno[2,3-d]pyrimidin-7-yl}carbonyl)azetidin-3-yl]carbamate (prepared according to example 54) were transformed in analogy to intermediate example 1g at 50°C overnight to give after working up and purification 22 mg (29%) of the title compound.

1H-NMR (400 MHz, DMSO-d6). δ [ppm]= 1.78 (1 H), 2.09 (1 H), 2.77 (1 H), 2.91 (2H), 3.08- 3.32 (4H), 3.49 (1 H), 3.70 (1 H), 3.82 (1 H), 4.02 (1 H), 4.37 (1 H), 6.65 (1 H), 7.55 (1 H), 8.07 (1 H), 8.31 (1 H), 8.41 (1 H), 8.73 (1 H)

Example 56

[(3R)-3,4-dimethylpiperazin-1 -yl]{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]- -tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}methanone

200 mg (506 pmol) (7S)-4-[(4-methoxypyrazolo[1 , 5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using (2R)-1 ,2- dimethylpiperazine dihydrochloride to give after working up and purification 218 mg (83%) of the title compound.

1 H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.00+1.02 (3H), 1.75-2.15 (4H), 2.19 (3H), 2.43- 2.53 (1 H), 2.72-3.05 (4H), 3.18-3.31 (3H), 3.87+3.92 (1 H), 4.01 (3H), 4.14+4.21 (1 H), 6.72 (1 H), 7.83 (1 H), 7.98 (1 H), 8.18 (1 H), 8.44 (1 H), 8.52 (1 H). Example 57 [(3S)-3,4-dimethylpiperazin-1 -yl]{(7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]- -tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}methanone

200 mg (506 pmol) (7S)-4-[(4-methoxypyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8- tetra ydro[1]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 7a) were transformed in analogy to example 1 using (2S)-1 ,2- dimethylpiperazine dihydrochloride to give after working up and purification 122 mg (47%) of the title compound.

H-NMR (400 MHz, DMSO-d6), δ [ppm]= 1.00+1.02 (3H), 1.78-2.15 (4H), 2.18+2.20 (3H), 2.46 (1 H), 2.72-3.04 (4H), 3.16-3.29 (3H), 3.89 (1 H), 4.00-4.03 (3H), 4.15+4.25 (1 H), 6.72 (1 H), 7.83 (1 H), 7.98 (1 H), 8.18 (1 H), 8.44 (1 H), 8.52 (1 H).

Example 58

{(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(3 )-3,4-dimethylpiperazin-1 -

200 mg (500 pmol) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1 ]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 1 a) were transformed in analogy to example 1 using (2R)-1 ,2- dimethylpiperazine dihydrochloride to give after working up and purification 198 mg (76%) of the title compound.

H-NMR (400 MHz, DMSO-d6), δ [ppm]= 0.99+1.02 (3H), 1.75-2.14 (4H), 2.19 (3H), 2.47 (1 H), 2.70-3.06 (4H), 3.17-3.30 (3H), 3.86+3.92 (1 H), 4.14-4.21 (1 H), 6.65 (1 H), 7.54 (1 H), 8.07 (1 H), 8.31 (1 H), 8.41 (1 H), 8.73 (1 H).

Examp!e 59

{(7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5, 6,7,8- tetrahydro[1]benzothieno[2,3-d]pyrimidin-7-yl}[(3S)-3,4-dime thylpiperazin-1- yl]methanone

200 mg (500 pmol) (7S)-4-[(4-chloropyrazolo[1 ,5-a]pyridin-5-yl)amino]-5,6,7,8- tetrahydro[1 ]benzothieno[2,3-d]pyrimidine-7-carboxylic acid (prepared according to intermediate example 1 a) were transformed in analogy to example 1 using (2S)-1 ,2- dimethylpiperazine dihydrochloride to give after working up and purification 136 mg (55%) of the title compound.

1 H-N R (400 MHz, DMSO-d6). δ [ppm]= 1.00+1.02 (3H), 1.74-2.14 (4H), 2.18+2.19 (3H), 2.46 (1 H), 2.72-3.06 (4H), 3.17-3.30 (3H), 3.89 (1 H), 4.14+4.25 (1 H), 6.66 (1 H), 7.54 (1 H), 8.08 (1 H), 8.31 (1 H), 8.42 (1 H), 8.74 (1 H).

Pharmaceutical compositions of the compounds of the invention

This invention also relates to pharmaceutical compositions containing one or more compounds of the present invention. These compositions can be utilised to achieve the desired pharmacological effect by administration to a patient in need thereof. A patient, for the purpose of this invention, is a mammal, including a human, in need of treatment for the particular condition or disease. Therefore, the present invention includes pharmaceutical compositions that are comprised of a pharmaceutically acceptable carrier and a

pharmaceutically effective amount of a compound, or salt thereof, of the present invention. A pharmaceutically acceptable carrier is preferably a carrier that is relatively non-toxic and innocuous to a patient at concentrations consistent with effective activity of the active ingredient so that any side effects ascribable to the carrier do not vitiate the beneficial effects of the active ingredient. A pharmaceutically effective amount of compound is preferably that amount which produces a result or exerts an influence on the particular condition being treated. The compounds of the present invention can be administered with

pharmaceutically-acceptable carriers well known in the art using any effective conventional dosage unit forms, including immediate, slow and timed release preparations, orally, parenterally, topically, nasally, ophthalmically, optically, sublingually, rectally, vaginally, and the like.

For oral administration, the compounds can be formulated into solid or liquid preparations such as capsules, pills, tablets, troches, lozenges, melts, powders, solutions, suspensions, or emulsions, and may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions. The solid unit dosage forms can be a capsule that can be of the ordinary hard- or soft-shelled gelatine type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and corn starch.

In another embodiment, the compounds of this invention may be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatine, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, gum tragacanth, acacia, lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid, or magnesium, calcium or zinc stearate, dyes, colouring agents, and flavouring agents such as peppermint, oil of wintergreen, or cherry flavouring, intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient. Suitable excipients for use in oral liquid dosage forms include dicalcium phosphate and diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both.

Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example those sweetening, flavouring and colouring agents described above, may also be present.

The pharmaceutical compositions of this invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil such as liquid paraffin or a mixture of vegetable oils. Suitable emulsifying agents may be (1 ) naturally occurring gums such as gum acacia and gum tragacanth, (2) naturally occurring phosphatides such as soy bean and lecithin, (3) esters or partial esters derived form fatty acids and hexitol anhydrides, for example, sorbitan monooleate, (4) condensation products of said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavouring agents.

Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil such as, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent such as, for example, beeswax, hard paraffin, or cetyl alcohol. The suspensions may also contain one or more preservatives, for example, ethyl or n-propyl p-hydroxybenzoate ; one or more colouring agents ; one or more flavouring agents ; and one or more sweetening agents such as sucrose or saccharin.

Syrups and elixirs may be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, and preservative, such as methyl and propyl parabens and flavouring and colouring agents.

The compounds of this invention may also be administered parenterally, that is,

subcutaneously, intravenously, intraocularly, intrasynovially, intramuscularly, or

interperitoneally, as injectable dosages of the compound in preferably a physiologically acceptable diluent with a pharmaceutical carrier which can be a sterile liquid or mixture of liquids such as water, saline, aqueous dextrose and related sugar solutions, an alcohol such as ethanol, isopropanol, or hexadecyl alcohol, glycols such as propylene glycol or polyethylene glycol, glycerol ketals such as 2,2-dimethyl-1 ,1 -dioxolane-4-methanol, ethers such as poly(ethylene glycol) 400, an oil, a fatty acid, a fatty acid ester or, a fatty acid glyceride, or an acetylated fatty acid glyceride, with or without the addition of a

pharmaceutically acceptable surfactant such as a soap or a detergent, suspending agent such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or

carboxymethylcellulose, or emulsifying agent and other pharmaceutical adjuvants.

Illustrative of oils which can be used in the parenteral formulations of this invention are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, sesame oil, cottonseed oil, corn oil, olive oil, petrolatum and mineral oil. Suitable fatty acids include oleic acid, stearic acid, isostearic acid and myristic acid. Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate. Suitable soaps include fatty acid alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamine acetates ; anionic detergents, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates ; non-ionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and poly(oxyethylene-oxypropylene)s or ethylene oxide or propylene oxide copolymers ; and amphoteric detergents, for example, alkyl-beta-aminopropionates, and 2-alkylimidazoline quarternary ammonium salts, as well as mixtures.

The parenteral compositions of this invention will typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimise or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile-lipophile balance (HLB) preferably of from about 12 to about 17. The quantity of surfactant in such formulation preferably ranges from about 5% to about 15% by weight. The surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB. Illustrative of surfactants used in parenteral formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.

The pharmaceutical compositions may be in the form of sterile injectable aqueous suspensions. Such suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents such as, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia ; dispersing or wetting agents which may be a naturally occurring phosphatide such as lecithin, a condensation product of an alkylene oxide with a fatty acid, for example, polyoxyethylene stearate, a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example,

heptadeca-ethyleneoxycetanol, a condensation product of ethylene oxide with a partial ester derived form a fatty acid and a hexitol such as polyoxyethylene sorbitol monooleate, or a condensation product of an ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride, for example polyoxyethylene sorbitan monooleate.

The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Diluents and solvents that may be employed are, for example, water, Ringer's solution, isotonic sodium chloride solutions and isotonic glucose solutions. In addition, sterile fixed oils are conventionally employed as solvents or suspending media. For this purpose, any bland, fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid can be used in the preparation of injectables.

A composition of the invention may also be administered in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are, for example, cocoa butter and polyethylene glycol.

Another formulation employed in the methods of the present invention employs transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art (see, e.g., US Patent No. 5,023,252, issued June 1 1 , 1991 ,

incorporated herein by reference). Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.

Controlled release formulations for parenteral administration include liposomal, polymeric microsphere and polymeric gel formulations that are known in the art.

It may be desirable or necessary to introduce the pharmaceutical composition to the patient via a mechanical delivery device. The construction and use of mechanical delivery devices for the delivery of pharmaceutical agents is well known in the art. Direct techniques for, for example, administering a drug directly to the brain usually involve placement of a drug delivery catheter into the patient's ventricular system to bypass the blood-brain barrier. One such implantable delivery system, used for the transport of agents to specific anatomical regions of the body, is described in US Patent No. 5,01 1 ,472, issued April 30, 1991 . The compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired. Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized.

Such ingredients and procedures include those described in the following references, each of which is incorporated herein by reference: Powell, M.F. et al., "Compendium of Excipients for Parenteral Formulations" PDA Journal of Pharmaceutical Science & 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 propellants (examples include but are not limited to carbon dioxide, CCI2F2.

F2CIC-CCIF2 and CCIFs) 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 di hydrate) carrying agents (examples include but are not limited to acacia syrup, aromatic syrup, aromatic elixir, cherry syrup, cocoa syrup, orange syrup, syrup, corn oil, mineral oil, peanut oil, sesame oil, bacteriostatic sodium chloride injection and bacteriostatic water for injection) chelating agents (examples include but are not limited to edetate disodium and edetic acid) colourants (examples include but are not limited to FD&C Red No. 3, FD&C Red No. 20, FD&C Yellow No. 6, FD&C Blue No. 2, D&C Green No. 5, D&C Orange No. 5, D&C Red No. 8, caramel and ferric oxide red) ; clarifying agents (examples include but are not limited to bentonite) ; emulsifying agents (examples include but are not limited to acacia, cetomacrogol, cetyl alcohol, glyceryl monostearate, lecithin, sorbitan monooleate, polyoxyethylene 50

monostearate) ; encapsulating agents (examples include but are not limited to gelatin and cellulose acetate phthalate) flavourants (examples include but are not limited to anise oil, cinnamon oil, cocoa, menthol, orange oil, peppermint oil and vanillin) ; humectants (examples include but are not limited to glycerol, propylene glycol and sorbitol) ; levigating agents (examples include but are not limited to mineral oil and glycerin) ; oils (examples include but are not limited to arachis oil, mineral oil, olive oil, peanut oil, sesame oil and vegetable oil) ; ointment bases (examples include but are not limited to lanolin, hydrophilic ointment, polyethylene glycol ointment, petrolatum, hydrophilic petrolatum, white ointment, yellow ointment, and rose water ointment) ; penetration enhancers (transdermal delivery) (examples include but are not limited to monohydroxy or polyhydroxy alcohols, mono-or polyvalent alcohols, saturated or unsaturated fatty alcohols, saturated or unsaturated fatty esters, saturated or unsaturated dicarboxylic acids, essential oils, phosphatidyl derivatives, cephalin, terpenes, amides, ethers, ketones and ureas) plasticizers (examples include but are not limited to diethyl phthalate and glycerol) ; solvents (examples include but are not limited to ethanol, corn oil, cottonseed oil, glycerol, isopropanol, mineral oil, oleic acid, peanut oil, purified water, water for injection, sterile water for injection and sterile water for irrigation) ; stiffening agents (examples include but are not limited to cetyl alcohol, cetyl esters wax, microcrystalline wax, paraffin, stearyl alcohol, white wax and yellow wax) ; suppository bases (examples include but are not limited to cocoa butter and polyethylene glycols (mixtures)) ; surfactants (examples include but are not limited to benzalkonium chloride, nonoxynol 10, oxtoxynol 9, polysorbate 80, sodium lauryl sulfate and sorbitan mono-palmitate) ; suspending agents (examples include but are not limited to agar, bentonite, carbomers, carboxymethyiceliulose sodium, hydroxyethyl cellulose, hydroxypropyi 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,

carboxymethyiceliulose 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,

carboxymethyiceliulose 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.

Lyophilised powder for IV administration: A sterile preparation can be prepared with (i) 100 - 1000 mg of the desired compound of this invention as a lyophilised powder, (ii) 32- 327 mg/mL sodium citrate, and (iii) 300 - 3000 mg Dextran 40. The formulation is reconstituted with sterile, injectable saline or dextrose 5% to a concentration of 10 to 20 mg/mL, which is further diluted with saline or dextrose 5% to 0.2 - 0.4 mg/mL, and is administered either IV bolus or by IV infusion over 15 - 60 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.

Combination therapies

The term "combination" in the present invention is used as known to persons skilled in the art and may be present as a fixed combination, a non-fixed combination or kit-of-parts.

A "fixed combination" in the present invention is used as known to persons skilled in the art and is defined as a combination wherein the said first active ingredient and the said second active ingredient are present together in one unit dosage or in a single entity. One example of a "fixed combination" is a pharmaceutical composition wherein the said first active ingredient and the said second active ingredient are present in admixture for simultaneous administration, such as in a formulation. Another example of a "fixed combination" is a pharmaceutical combination wherein the said first active ingredient and the said second active ingredient are present in one unit without being in admixture.

A non-fixed combination or "kit-of-parts" in the present invention is used as known to persons skilled in the art and is defined as a combination wherein the said first active ingredient and the said second active ingredient are present in more than one unit. One example of a non-fixed combination or kit-of-parts is a combination wherein the said first active ingredient and the said second active ingredient are p-— ^iparately. The components of the non-fixed combination or kit-of-parts may be administered separately, sequentially, simultaneously, concurrently or chronologically staggered.

The compounds of this invention can be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutical agents where the combination causes no unacceptable adverse effects. The present invention relates also to such combinations. For example, the compounds of this invention can be combined with known chemotherapeutic agents or anti-cancer agents, e.g. anti-hyper-proliferative or other indication agents, and the like, as well as with admixtures and combinations thereof. Other indication agents include, but are not limited to, anti-angiogenic agents, mitotic inhibitors, alkylating agents, anti-metabolites, DNA-intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzyme inhibitors, topoisomerase inhibitors, proteasome inhibitors, biological response modifiers, anti-hormones or agents used for the treatment of inflammatory diseases or pain disorders. The terms "chemotherapeutic agent" and anti-cancer agent", include but are not limited to 131 l-chTNT, abarelix, abiraterone, aclarubicin, aldesleukin, alemtuzumab, alitretinoin, altretamine, aminoglutethimide, amrubicin, amsacrine, anastrozole, arglabin, arsenic trioxide, asparaginase, azacitidine, basiliximab, BAY 80-6946, BAY 1000394, BAY 86-9766 (RDEA 1 19), belotecan, bendamustine, bevacizumab, bexarotene, bicalutamide, bisantrene, bleomycin, bortezomib, buserelin, busulfan, cabazitaxel, calcium folinate, calcium

levofolinate, capecitabine, carboplatin, carmofur, carmustine, catumaxomab, celecoxib, celmoleukin, cetuximab, chlorambucil, chlormadinone, chlormethine, cisplatin, cladribine, clodronic acid, clofarabine, crisantaspase, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, darbepoetin alfa, dasatinib, daunorubicin, decitabine, deforolimus, degarelix, denileukin diftitox, denosumab, deslorelin, dibrospidium chloride, docetaxel, doxifluridine, doxorubicin, doxorubicin + estrone, eculizumab, edrecolomab, elliptinium acetate, eltrombopag, endostatin, enocitabine, enzastaurin, epirubicin, epitiostanol, epoetin alfa, epoetin beta, eptaplatin, eribulin, erlotinib, estradiol, estramustine, etoposide, everolimus, exemestane, fadrozole, filgrastim, fludarabine, fluorouracil, flutamide, formestane, fotemustine, fulvestrant, gallium nitrate, ganirelix, gefitinib, gemcitabine, gemtuzumab, glutoxim, goserelin, histamine dihydrochloride, histrelin, hydroxycarbamide, 1-125 seeds, ibandronic acid, ibritumomab tiuxetan, idarubicin, ifosfamide, imatinib, imiquimod, improsulfan, interferon alfa, interferon beta, interferon gamma, ipilimumab, irinotecan, ixabepilone, lanreotide, lapatinib, larotaxel, lenalidomide, lenograstim, lentinan, letrozole, leuprorelin, levamisole, lisuride, lobaplatin, lomustine, lonidamine, masoprocol, medroxyprogesterone, megestrol, melphalan, mepitiostane, mercaptopurine, methotrexate, methoxsalen, Methyl aminolevulinate, methyltestosterone, mifamurtide, miltefosine, miriplatin, mitobronitol, mitoguazone, mitolactol, mitomycin, mitotane, mitoxantrone, nedaplatin, nelarabine, nilotinib, nilutamide, nimotuzumab, nimustine, nitracrine, novolimus, ofatumumab, omeprazole, oprelvekin, oxaliplatin, p53 gene therapy, paclitaxel, palifermin, palladium-103 seed, pamidronic acid, panitumumab, pazopanib, pegaspargase,

PEG-epoetin beta (methoxy PEG-epoetin beta), pegfilgrastim, peginterferon alfa-2b, pemetrexed, pentazocine, pentostatin, peplomycin, perfosfamide, perifosine, picibanil, pirarubicin, plerixafor, plicamycin, poliglusam, polyestradiol phosphate, polysaccharide-K, porfimer sodium, pralatrexate, prednimustine, procarbazine, quinagolide, raloxifene, raltitrexed, ranimustine, rapamycin, razoxane, regorafenib, risedronic acid, rituximab, romidepsin, romiplostim, sagopilone, sargramostim, selumetinib, sipuleucel-T, sizofiran, sobuzoxane, sodium glycididazole, sorafenib, streptozocin, sunitinib, talaporfin,

tamibarotene, tamoxifen, tasonermin, teceleukin, tegafur, tegafur + gimeracil + oteracil, temoporfin. temozolomide, temsirolimus, teniposide, testosterone, tetrofosmin, thalidomide, thiotepa, thymalfasin, tioguanine, tocilizumab, topotecan, toremifene, tositumomab, trabectedin, trametinib, trastuzumab, treosulfan, tretinoin, triciribine, trilostane, triptorelin, trofosfamide, tryptophan, ubenimex, valrubicin, vandetanib, vapreotide, vemurafenib, vinblastine, vincristine, vindesine, vinflunine, vinorelbine, vorinostat, vorozole, yttrium-90 glass microspheres, zinostatin, zinostatin stimaiamer, zoledronic acid, zorubicin, zotarolimus, ARRY-162, ARRY-300, ARRY-704, AS-703026, AZD-5363, AZD-8055, BEZ-235, BGT-226, BKM-120, BYL-719, CAL-101 , CC-223, CH-5132799, E-6201. GDC-0032, GDC-0068, GDC-0623, GDC-0941 , GDC-0973, GDC-0980, GSK-21 10183, GSK-2126458,

GSK-2141795, INK128, MK-2206, OSI-027, PF-04691502, PF-05212384, PX-866, RG-7167, RO-4987655, RO-5126766, TAK-733, UCN-01. WX-554, XL- 147, XL-765, ZSTK-474. The terms "chemotherapeutic agent" and anti-cancer agent", also include protein

therapeutics such as an interferon (e.g., interferon .alpha., .beta., or .gamma.) supraagonistic monoclonal antibodies, Tuebingen, TRP-1 protein vaccine, Colostrinin, anti-FAP antibody, YH-16, gemtuzumab, infliximab, cetuximab, trastuzumab, denileukin diftitox, rituximab, thymosin alpha 1 , bevacizumab, mecasermin, mecasermin rinfabate, oprelvekin,

natalizumab, rhMBL, MFE-CP1 + ZD-2767-P, ABT-828, ErbB2-specific immunotoxin,

SGN-35, MT-103, rinfabate, AS-1402, B43-genistein, L-19 based radioimmunotherapeutics, AC-9301 , NY-ESO-1 vaccine, IMC-1 C1 1 , CT-322, rhCCI O, r(m)CRP, MORAb-009, aviscumine, MDX-1307, Her-2 vaccine, APC-8024, NGR-hTNF, rhH1.3, IGN-31 1 ,

Endostatin, volociximab, PRO-1762, lexatumumab, SGN-40, pertuzumab, EMD-273063, L19-IL-2 fusion protein, PRX-321 , CNTO-328, MDX-214, tigapotide, CAT-3888,

labetuzumab, alpha-particle-emitting radioisotope-llinked lintuzumab, EM-1421 , HyperAcute vaccine, tucotuzumab celmoleukin, galiximab, HPV-16-E7, Javelin - prostate cancer, Javelin - melanoma, NY-ESO-1 vaccine, EGF vaccine, CYT-004-MelQbG10, WT1 peptide, oregovomab, ofatumumab, zalutumumab, cintredekin besudotox, WX-G250, Albuferon, aflibercept, denosumab, vaccine, CTP-37, efungumab, or 131 l-chTNT-1/B.

The terms "chemotherapeutic agent" and "anti-cancer agent", also include monoclonal antibodies useful as the protein therapeutic such as muromonab-CD3, abciximab, edrecolomab, daclizumab, gentuzumab, alemtuzumab, ibritumomab, cetuximab, bevicizumab, efalizumab, adalimumab, omalizumab, muromomab-CD3, rituximab, daclizumab, trastuzumab, palivizumab, basiliximab, and infliximab.

Generally, the use of cytotoxic and/or cytostatic agents in combination with a compound or composition of the present invention will serve to:

(1 ) yield better efficacy in reducing the growth of a tumor or even eliminate the tumor as compared to administration of either agent alone,

(2) provide for the administration of lesser amounts of the administered chemotherapeutic agents,

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

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

especially humans,

(5) provide for a higher response rate among treated patients,

(6) provide for a longer survival time among treated patients compared to standard

chemotherapy treatments,

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

(8) yield efficacy and tolerability results at least as good as those of the agents used alone, compared to known instances where other cancer agent combinations produce antagonistic effects.

Beyond the "chemotherapeutic agent" and "anti cancer agent" the invention can be combined with further "anti-inflammatory" and 'anti-pain agents" which include but are not limited to abatacept, or anti-bacterial agents (e.g. penicilline, vancomycin, ciprofloxacin), anti-viral agents (e.g. aciclovir, oseltamivir), anti-mycotic agents (e.g. naftifine, nystatin), azathioprine, belimumab, corticosteroids (e.g. prednisor ~ ' ~ 3olone, methylprednisolone, hydrocortisone, betamethasone), cyclophosphamide, IgE antibody, immunoglobulin and gammaglobuline, IL-1 inhibitors (e.g. anakinra, canakinumab, rilonacept),

"immunomodulatory and immunosuppressive agents " like cyclosporine, mercaptopurine, Methotrexate®; interferon including beta-interferon (IFN beta-1 a: Avonex® and IFN beta-1 b: Betaferon®), Jak/STAT inhibitors (e.g. tofacitinib, baricitinib, GLPG0634), leflunomide, mycophenolic acid, nonsteroidal anti-inflammatory drugs (NSAIDS) (e.g. ibuprofen, naproxen, etodolac, celecoxib, colchicine), paracetamol, phosphodiesterase-inhibitor (e.g. apremilast, roflumilast), rapamycin, rituximab, sulfasalazine, tacrolimus and TNF-antagonist (e.g. Humira®, etanercept, infliximab). In addition, combination also includes ACE (angiotensin-converting-enzyme) inhibitors (e.g. benazepril), acetylsalicylic acid, acetylcholinesterase inhibitors (e.g. donepezil, rivastigmine, galantamine, tacrine), anticholinergic agents (e.g. trihexyphenidyle, glycopyrronium bromid), anticonvulsant agents (e.g. gabapentin), anti-diarrhoeal drug (e.g. loperamide or laxatives), antileukotriene agents (e.g. montelukast), beta blocker (e.g. metoprolol), beta2-adrenergic agonists (e.g. salbutamol), calcium channel blockers (e.g. nifedipine), chloroquine, COMT (Catechol-O- ethyltransferase)-inhibitors (e.g. entacapone), diuretics (e.g.

hydrochlorothiazide), dopamine agonists (e.g. ropinrole, pramipexole, bromocriptine), efalizumab, fingolimod, glatiramer acetate, glibenclamide, insulin therapy, L- DOPA/Carbidopa (L-3,4-Dihydroxyphenylalanin), MAO-B (monoamine oxidase B) inhibitors (e.g. selegiline), mesalazine, metformin, methylxanthine drugs (e.g. theophylline), mitoxantrone, natalizumab, NMDA (N-Methyl-D-Aspartat) receptor antagonists (e.g.

amantadine, memantine), probiotics (e.g. mutaflor, VSL#3®, Lactobacillus GG, Lactobacillus plantarum, L. acidophilus, L. casei, Bifidobacterium infantis 35624, Enterococcus fecium SF68, Bifidobacterium longum), statin (e.g. simvastatin), sulfonylureas (e.g. tolnutamide, glimepiride), urea and vitamin-D analoga (e.g. calcipotriol, calcitriol, tacalcitol).

Methods of Sensitizing Ceils to Radiation

In a distinct embodiment of the present invention, a compound of the present invention may be used to sensitize a cell to radiation. That is, treatment of a cell with a compound of the present invention prior to radiation treatment of the cell renders the cell more susceptible to DNA damage and cell death than the cell would be in the absence of any treatment with a compound of the invention. In one aspect, the cell is treated with at least one compound of the invention.

Thus, the present invention also provides a method of killing a cell, wherein a cell is administered one or more compounds of the invention in combination with conventional radiation therapy. The present invention also provides a method of rendering a cell more susceptible to cell death, wherein the cell is treated with one or more compounds of the invention prior to the treatment of the cell to cause or induce cell death. In one aspect, after the cell is treated with one or more compounds of the invention, the cell is treated with at least one compound, or at least one method, or a combination thereof, in order to cause DNA damage for the purpose of inhibiting the function of the normal cell or killing the cell.

In one embodiment, a cell is killed by treating the cell with at least one DNA damaging agent. That is, after treating a cell with one or more compounds of the invention to sensitize the cell to cell death, the cell is treated with at least one DNA damaging agent to kill the cell. DNA damaging agents useful in the present invention include, but are not limited to,

chemotherapeutic agents (e.g., cisplatinum), ionizing radiation (X-rays, ultraviolet radiation), carcinogenic agents, and mutagenic agents.

In another embodiment, a cell is killed by treating the cell with at least one method to cause or induce DNA damage. Such methods include, but are not limited to, activation of a cell signalling pathway that results in DNA damage when the pathway is activated, inhibiting of a cell signalling pathway that results in DNA damage when the pathway is inhibited, and inducing a biochemical change in a cell, wherein the change results in DNA damage. By way of a non-limiting example, a DNA repair pathway in a cell can be inhibited, thereby preventing the repair of DNA damage and resulting in an abnormal accumulation of DNA damage in a cell.

In one aspect of the invention, a compound of the invention is administered to a cell prior to the radiation or other induction of DNA damage in the cell. In another aspect of the invention, a compound of the invention is administered to a cell concomitantly with the radiation or other induction of DNA damage in the cell. In yet another aspect of the invention, a compound of the invention is administered to a cell immediately after radiation or other induction of DNA damage in the cell has begun.

In another aspect, the cell is in vitro. In another embodiment, the cell is in vivo.

As mentioned supra, the compounds of the present invention have surprisingly been found to effectively inhibit MKNK1 and may therefore be used for the treatment or prophylaxis of diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, or diseases which are accompanied with uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, particularly in which the uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses is mediated by MKNK1 , such as, for example, haematological tumours, solid tumours, and/or metastases thereof, e.g. leukaemias and myelodysplastic syndrome, malignant lymphomas, head and neck tumours including brain tumours and brain metastases, tumours of the thorax including non-small cell and small cell lung tumours, gastrointestinal tumours, endocrine tumours, mammary and other gynaecological tumours, urological tumours including renal, bladder and prostate tumours, skin tumours, and sarcomas, and/or metastases thereof, or pancreatitis.

In accordance with another aspect therefore, the present invention covers a compound of general formula (I), or a stereoisomer, a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, as described and defined herein, for use in the treatment or prophylaxis of a disease, as mentioned supra.

Another particular aspect of the present invention is therefore the use of a compound of general formula (I), described supra, or a stereoisomer, a tautomer, an N -oxide, a hydrate, a solvate, or a salt thereof, particularly a pharmaceutically acceptable salt thereof, or a mixture of same, for the prophylaxis or treatment of a disease.

Another particular aspect of the present invention is therefore the use of a compound of general formula (I) described supra for manufacturing a pharmaceutical composition for the treatment or prophylaxis of a disease.

The diseases referred to in the two preceding paragraphs are diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, or diseases which are accompanied with uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, particularly in which the uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses is mediated by MKNK1 , such as, for example, haematological tumours, solid tumours, and/or metastases thereof, e.g. leukaemias and myelodysplastic syndrome, malignant lymphomas, head and neck tumours including brain tumours and brain metastases, tumours of the thorax including non-small cell and small cell lung tumours, gastrointestinal tumours, endocrine tumours, mammary and other gynaecological tumours, urological tumours including renal, bladder and prostate tumours, skin tumours, and sarcomas, and/or metastases thereof. The term "inappropriate" within the context of the present invention, in particular in the context of "inappropriate cellular immune responses, or inappropriate cellular inflammatory responses", as used herein, is to be understood as preferably meaning a response which is less than, or greater than normal, and which is associated with, responsible for, or results in, the pathology of said diseases.

Preferably, the use is in the treatment or prophylaxis of diseases, wherein the diseases are haemotological tumours, solid tumours and/or metastases thereof.

Method of treating hyper-proliferative disorders

The present invention relates to a method for using the compounds of the present invention and compositions thereof, to treat mammalian hyper-proliferative disorders. Compounds can be utilized to inhibit, block, reduce, decrease, etc., cell proliferation and/or cell division, and/or produce apoptosis. This method comprises administering to a mammal in need thereof, including a human, an amount of a compound of this invention, or a

pharmaceutically acceptable salt, isomer, polymorph, metabolite, hydrate, solvate or ester thereof ; etc. which is effective to treat the disorder. Hyper-proliferative disorders include but are not limited, e.g., psoriasis, keloids, and other hyperplasias affecting the skin, benign prostate hyperplasia (BPH), solid tumours, such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid and their distant metastases. Those disorders also include lymphomas, sarcomas, and leukaemias.

Examples of breast cancer include, but are not limited to invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ.

Examples of cancers of the respiratory tract include, but are not limited to small-cell and non-small-cell lung carcinoma, as well as bronchial adenoma and pleuropulmonary blastoma.

Examples of brain cancers include, but are not limited to brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal tumour.

Tumours of the male reproductive organs include, but are not limited to prostate and testicular cancer. Tumours of the female reproductive organs include, but are not limited to endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus. Tumours of the digestive tract include, but are not limited to anal, colon, colorectal, oesophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers. Tumours of the urinary tract include, but are not Iimited to bladder, penile, kidney, renal pelvis, ureter, urethral and human papillary renal cancers.

Eye cancers include, but are not Iimited to intraocular melanoma and retinoblastoma.

Examples of liver cancers include, but are not Iimited to hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma.

Skin cancers include, but are not Iimited to squamous cell carcinoma, Kaposi's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer.

Head-and-neck cancers include, but are not Iimited to laryngeal, hypopharyngeal, nasopharyngeal, oropharyngeal cancer, lip and oral cavity cancer and squamous cell.

Lymphomas include, but are not Iimited to AIDS-related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Burkitt lymphoma, Hodgkin's disease, and lymphoma of the central nervous system.

Sarcomas include, but are not Iimited to sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma.

Leukemias include, but are not Iimited to acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia.

These disorders have been well characterized in humans, but also exist with a similar etiology in other mammals, and can be treated by administering pharmaceutical

compositions of the present invention.

The term "treating" or "treatment" as stated throughout this document is used conventionally, e.g., the management or care of a subject for the purpose of combating, alleviating, reducing, relieving, improving the condition of, efc, of a disease or disorder, such as a carcinoma. Methods of treating kinase disorders

The present invention also provides methods for the treatment of disorders associated with aberrant mitogen extracellular kinase activity, including, but not Iimited to stroke, heart failure, hepatomegaly, cardiomegaly, diabetes, Alzheimer's disease, cystic fibrosis, symptoms of xenograft rejections, septic shock or asthma.

Effective amounts of compounds of the present invention can be used to treat such disorders, including those diseases (e.g., cancer) mentioned in the Background section above. Nonetheless, such cancers and other diseases can be treated with compounds of the present invention, regardless of the mechanism of action and/or the relationship between the kinase and the disorder.

The phrase "aberrant kinase activity" or "aberrant serin threonin kinase activity," includes any abnormal expression or activity of the gene encoding the kinase or of the polypeptide it encodes. Examples of such aberrant activity, include, but are not limited to, over-expression of the gene or polypeptide ; gene amplification ; mutations which produce

constitutively-active or hyperactive kinase activity ; gene mutations, deletions, substitutions, additions, etc. The present invention also provides for methods of inhibiting a kinase activity, especially of mitogen extracellular kinase, comprising administering an effective amount of a compound of the present invention, including salts, polymorphs, metabolites, hydrates, solvates, prodrugs (e.g.: esters) thereof, and diastereoisomeric forms thereof. Kinase activity can be inhibited in cells (e.g., in vitro), or in the cells of a mammalian subject, especially a human patient in need of treatment.

Methods of treating pain-associated diseases and gynaecological disorders.

The present invention also provides methods for the treatment or prophylaxis of inflammation and pain-associated diseases.

In particular aspect of the invention as reported above a compound of formula (I), (la) or (lb) is for the treatment of pain syndromes including acute, chronic, inflammatory and neuropathic pain, preferably inflammatory pain, surgical pain, visceral pain, dental pain, premenstrual pain, gynaecological disease, preferably dysmenorrhea, dyspareunia or endometriosis, adenomyosis, endometriosis-associated pain, or other endometriosis- associated symptoms, wherein said symptoms are in particular endometriosis-associated dysmenorrhea, dyspareunia, dysuria, or dyschezia, pain associated with fibrotic diseases, central pain, pain due to burning mouth syndrome, pain due to burns, pain due to migraine, cluster headaches, pain due to nerve injury, pain due to neuritis, neuralgias, pain due to poisoning, pain due to ischemic injury, pain due to interstitial cystitis, cancer pain, pain due to viral, parasitic or bacterial infections, pain due to traumatic nerve-injury, pain due to post- traumatic injuries (including fractures and sport injuries), pain due to trigeminal neuralgia, pain associated with small fiber neuropathy, pain associated with diabetic neuropathy, chronic lower back pain, phantom limb pain, pelvic pain syndrome, chronic pelvic pain, neuroma pain, complex regional pain syndrome, pain associated with gastrointestinal distension, chronic arthritic pain and related neuralgias, and pain associated with cancer, pain associated with chemotherapy, HIV and HIV treatment-induced neuropathy; and pain associated with diseases or disorders selected from the group consisting of hyperalgesia, allodynia, irritable bowel syndrome.

In addition, the present invention is for the use of the treatment and prevention of inflammatory diseases including inflammatory bowel disease (ulcerative colitis and Crohn's disease), hyperaemia, sepsis, metabolic disorders, e.g. obesity, insulin resistance, diabetes mellitus type 1 and 2, metabolic endocrine disorder; metabolic syndrome: atherosclerosis, reperfusion injury, inflammatory bone resorption, inflammatory liver diseases, pulmonary fibrosis, acute respiratory distress syndrome, and intestinal polyposis, inflammatory skin diseases like psoriasis, pemphigus vulgaris, inflammatory eye disorders like non-infectious uveitis, primary and secondary autoimmune uveitis, VKH syndrome, anterior uveitis, intermediate uveitis, posterior uveitis, panuveitis, Behcet's disease and neuromyelitis optica ,fibrotic diseases like idiopathic pulmonary fibrosis, skin fibrosis, systemic sclerosis, autism disorders, liver diseases like nonalcoholic-, alcoholic- and toxic fatty liver disease, steatohepatitis, hepatic fibrosis; and cirrhosis; lung diseases like chronic obstructive pulmonary disease, asthma, pneumonia; neurodegenerative diseases like Parkinson's disease, Alzheimer's disease; stroke, postischemic brain injury, brain ischemia, posttraumatic brain injury, alopecia, acute coronary syndrome, myocardial infarction, autoimmune diseases like autoimmune encephalomyelitis, multiple sclerosis; arthritis (such as osteoarthritis and rheumatoid arthritis); , psoriatic arthritis, ankylosing spondylitis), psoriasis, lupus erythematosus (e.g. systemic lupus erythematosus, cutaneous lupus and neonatal lupus erythematosus); inflammatory, autoimmune and fibrotic kidney diseases (e.g.

glomerulonephritis, lupus nephritis, ANCA) interstitial cystitis, hypertrophy of the e.g. kidney, ischemia/reperfusion injury, allergic rhinitis, burn wound, osteoporosis viral and bacterial infections, chemotherapy-induced alopecia, cachexia induced for any reason, e.g. cancer, heart failure, etc., muscular atrophy, pancreatitis, schizophrenia, seizures, epilepsy, Fragile X syndrome, graft-versus-host disease, graft rejection, heart fibrosis, autoimmune myocardial disease, myocarditis, endocarditis, ischemia-reperfusion injury following e.g. myocardial infarction, hypertension, artherosclerosis, acute lung injury, ARDS, hypersensitivity pneumonitis, lung fibrosis, e.g. idiopathic pulmonary fibrosis, lymphocytic bronchiolitis e.g. after lung transplantation, dry eye, subfertility (e.g. associated with inflammatory conditions such as endometriosiso, or metabolic endocrine disorders).

Dose and administration

Based upon standard laboratory techniques known to evaluate compounds useful for the treatment of hyper-proliferative disorders and angiogenic disorders, by standard toxicity tests and by standard pharmacological assays for the determination of treatment of the conditions identified above in mammals, and by comparison of these results with the results of known medicaments that are used to treat these conditions, the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication. The amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.

The total amount of the active ingredient to be administered will generally range from about 0.001 mg/kg to about 200 mg/kg body weight per day, and preferably from about 0.01 mg/kg to about 20 mg/kg body weight per day. Clinically useful dosing schedules will range from one to three times a day dosing to once every four weeks dosing. In addition, "drug holidays" in which a patient is not dosed with a drug for a certain period of time, may be beneficial to the overall balance between pharmacological effect and tolerability. A unit dosage may contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered one or more times per day or less than once a day. The average daily dosage for administration by injection, including intravenous, intramuscular, subcutaneous and parenteral injections, and use of infusion techniques will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily rectal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily vaginal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily topical dosage regimen will preferably be from 0.1 to 200 mg administered between one to four times daily. The transdermal concentration will preferably be that required to maintain a daily dose of from 0.01 to 200 mg/kg. The average daily inhalation dosage regimen will preferably be from 0.01 to 100 mg/kg of total body weight.

Of course the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like. The desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.

Preferably, the diseases of said method are haematological tumours, solid tumour and/or metastases thereof. The compounds of the present invention can be used in particular in therapy and prevention, i.e. prophylaxis, of tumour growth and metastases, especially in solid tumours of all indications and stages with or without pre-treatment of the tumour growth. Methods of testing for a particular pharmacological or pharmaceutical property are well known to persons skilled in the art.

The example testing experiments described herein serve to illustrate the present invention and the invention is not limited to the examples given. Biological assays

Examples were tested in selected biological assays one or more times. When tested more than once, data are reported as either average values or as median values, wherein

• the average value, also referred to as the arithmetic mean value, represents the sum of the values obtained divided by the number of times tested, and

· the median value represents the middle number of the group of values when ranked in ascending or descending order. If the number of values in the data set is odd, the median is the middle value. If the number of values in the data set is even, the median is the arithmetic mean of the two middle values.

Examples were synthesized one or more times. When synthesized more than once, data from biological assays represent average values or median values calculated utilizing data sets obtained from testing of one or more synthetic batch.

MKNK1 kinase assay

MKNK1 -inhibitory activity of compounds of the present invention was quantified employing the MKNK1 TR-FRET assay as described in the following paragraphs.

A recombinant fusion protein of Glutathione-S-Transferase (GST, N-terminally) and human full-length MKNK1 (amino acids 1-424 and T344D of accession number BAA 19885.1 ), expressed in insect cells using baculovirus expression system and purified via glutathione sepharose affinity chromatography, was purchased from Carna Biosciences (product no 02-145) and used as enzyme. As substrate for the kinase reaction the biotinylated peptide biotin-Ahx-IKKRKLTRRKSLKG (C-terminus in amide form) was used which can be purchased e.g. form the company Biosyntan (Berlin-Buch, Germany).

For the assay 50 nL of a 10Ofold concentrated solution of the test compound in DMSO was pipetted into a black low volume 384well microtiter plate (Greiner Bio-One, Frickenhausen, Germany), 2 μΙ_ of a solution of MKNK1 in aqueous assay buffer [50 mM HEPES pH 7.5, 5 mM MgC , 1.0 mM dithiothreitol, 0.005% (v/v) Nonidet-P40 (Sigma)] was added and the mixture was incubated for 15 min at 22°C to allow pre-binding of the test compounds to the enzyme before the start of the kinase reac t; n Tho n the kinase reaction was started by the addition of 3 μΙ_ of a solution of adenosine-tri-phosphate (ATP, 16.7 μΜ => final cone, in the 5 μΙ_ assay volume is 10 μΜ) and substrate (0.1 μΜ => final cone, in the 5 sL assay volume is 0.06 μΜ) in assay buffer and the resulting mixture was incubated for a reaction time of 45 min at 22°C. The concentration of MKNK1 was adjusted depending of the activity of the enzyme lot and was chosen appropriate to have the assay in the linear range, typical concentrations were in the range of 0.05 μg/ml. The reaction was stopped by the addition of 5 μΙ_ of a solution of TR-FRET detection reagents (5 nM streptavidine-XL665 [Cisbio Bioassays, Codolet, France] and 1 nM anti-ribosomal protein S6 (pSer236)-antibody from Invitrogen [# 44921 G] and 1 nM LANCE EU-W1024 labeled ProteinG [Perkin-Elmer, product no. AD0071 ]) in an aqueous EDTA-solution (100 mM EDTA, 0.1 % (w/v) bovine serum albumin in 50 mM HEPES pH 7.5).

The resulting mixture was incubated for 1 h at 22 °C to allow the formation of complex between the phosphorylated biotinylated peptide and the detection reagents. Subsequently the amount of phosphorylated substrate was evaluated by measurement of the resonance energy transfer from the Eu-chelate to the streptavidine-XL. Therefore, the fluorescence emissions at 620 nm and 665 nm after excitation at 350 nm were measured in a TR-FRET reader, e.g. a Rubystar (BMG Labtechnologies, Offenburg, Germany) or a Viewlux

(Perkin-Elmer). The ratio of the emissions at 665 nm and at 622 nm was taken as the measure for the amount of phosphorylated substrate. The data were normalised (enzyme reaction without inhibitor = 0 % inhibition, all other assay components but no enzyme = 100 % inhibition). Usually the test compounds were tested on the same microtiterplate in 1 1 different concentrations in the range of 20 μΜ to 0.1 nM (20 μΜ, 5.9 μΜ, 1 .7 μΜ, 0.51 μΜ, 0.15 μΜ, 44 nM, 13 nM, 3.8 nM, 1 .1 nM, 0.33 nM and 0.1 nM, the dilution series prepared separately before the assay on the level of the 10Ofold concentrated solutions in DMSO by serial 1 :3.4 dilutions) in duplicate values for each concentration and ICso values were calculated by a 4 parameter fit using an inhouse software.

MKNK1 kinase high ATP assay

MKNK1 -inhibitory activity at high ATP of compounds of the present invention after their preincubation with MKNK1 was quantified employing the TR-FRET-based MKNK1 high ATP assay as described in the following paragraphs.

A recombinant fusion protein of Glutathione-S-Transferase (GST, N-terminally) and human full-length MKNK1 (amino acids 1 -424 and T344D of accession number BAA 19885.1 ), expressed in insect cells using baculovirus expression system and purified via glutathione sepharose affinity chromatography, was purchased from Carna Biosciences (product no 02-145) and used as enzyme. As substrate for the kinase reaction the biotinylated peptide biotin-Ahx-IKKRKLTRRKSLKG (C-terminus in amide form) was used, which can be purchased e.g. from the company Biosyntan (Berlin-Buch, Germany). For the assay 50 nl_ of a 10Ofold concentrated solution of the test compound in DMSO was pipetted into a black low volume 384well microtiter plate (Greiner Bio-One, Frickenhausen, Germany), 2 μΙ_ of a solution of MKNK1 in aqueous assay buffer [50 mM HEPES pH 7.5, 5 mM MgC , 1.0 mM dithiothreitol, 0.005% (v/v) Nonidet-P40 (Sigma)] was added and the mixture was incubated for 15 min at 22°C to allow pre-binding of the test compounds to the enzyme before the start of the kinase reaction. Then the kinase reaction was started by the addition of 3 μΙ_ of a solution of adenosine-tri-phosphate (ATP, 3.3 mM => final cone, in the 5 μΙ_ assay volume is 2 mM) and substrate (0.1 μΜ => final cone, in the 5 μΙ_ assay volume is 0.06 μΜ) in assay buffer and the resulting mixture was incubated for a reaction time of 30 min at 22°C. The concentration of MKNK1 was adjusted depending of the activity of the enzyme lot and was chosen appropriate to have the assay in the linear range, typical concentrations were in the range of 0.003 μg mL. The reaction was stopped by the addition of 5 μΙ_ of a solution of TR-FRET detection reagents (5 nM streptavidine-XL665 [Cisbio Bioassays, Codolet, France] and 1 nM anti-ribosomal protein S6 (pSer236)-antibody from Invitrogen [# 44921 G] and 1 nM LANCE EU-W1024 labeled ProteinG [Perkin-Elmer, product no. AD0071]) in an aqueous EDTA-solution (100 mM EDTA, 0.1 % (w/v) bovine serum albumin in 50 mM HEPES pH 7.5).

The resulting mixture was incubated for 1 h at 22°C to allow the formation of complex between the phosphorylated biotinylated peptide and the detection reagents. Subsequently the amount of phosphorylated substrate was evaluated by measurement of the resonance energy transfer from the Eu-chelate to the streptavidine-XL. Therefore, the fluorescence emissions at 620 nm and 665 nm after excitation at 350 nm were measured in a TR-FRET reader, e.g. a Rubystar (BMG Labtechnologies, Offenburg, Germany) or a Viewlux

(Perkin-Elmer). The ratio of the emissions at 665 nm and at 622 nm was taken as the measure for the amount of phosphorylated substrate. The data were normalised (enzyme reaction without inhibitor = 0 % inhibition, all other assay components but no enzyme = 100 % inhibition). Usually the test compounds were tested on the same microtiterplate in 1 1 different concentrations in the range of 20 μΜ to 0.1 nM (e.g. 20 μΜ, 5.9 μΜ, 1.7 μΜ, 0.51 μΜ, 0.15 μΜ, 44 nM, 13 nM, 3.8 nM, 1.1 nM, 0.33 nM and 0.1 nM, the dilution series prepared separately before the assay on the level of the 10Ofold concentrated solutions in DMSO by serial dilutions, the exact concentrations may vary depending on the pipettor used) in duplicate values for each concentration and IC50 values were calculated. Data are presented in Table 1. Table 1

: not yet determined MKNK 2 kinase high ATP assay

MKNK 2-inhibitory activity at high ATP of compounds of the present invention after their preincubation with MKNK 2 was quantified employing the TR-FRET-based MKNK 2 high ATP assay as described in the following paragraphs.

A recombinant fusion protein of Glutathione-S-Transferase (GST, N-terminally) and human full-length MKNK 2 (Genbank accession number NP_ 060042.2), expressed in insect cells using baculovirus expression system , purified via glutathione sepharose affinity

chromatography, and activated in vitro with MAPK12, was purchased from Invitrogen (product no PV5608) and used as enzyme. As substrate for the kinase reaction the biotinylated peptide biotin-Ahx-IKKRKLTRRKSLKG (C-terminus in amide form) was used which can be purchased e.g. form the company Biosyntan (Berlin-Buch, Germany).

For the assay 50 nl of a 10Ofold concentrated solution of the test compound in DMSO was pipetted into a black low volume 384well microtiter plate (Greiner Bio-One, Frickenhausen, Germany), 2 μΙ of a solution of MKNK 2 in aqueous assay buffer [50 mM HEPES pH 7.5, 5 mM MgC , 1.0 mM dithiothreitol, 0.005% (v/v) Nonidet-P40 (G-Biosciences, St. Louis, USA)] was added and the mixture was incubated for 15 min at 22°C to allow pre-binding of the test compounds to the enzyme before the start of the kinase reaction. Then the kinase reaction was started by the addition of 3 μΙ of a solution of adenosine-tri-phosphate (ATP,

3.3 mM => final cone, in the 5 μΙ assay volume is 2 mM) and substrate (0.1 μΜ => final cone, in the 5 μΙ assay volume is 0.06 μΜ) in assay buffer and the resulting mixture was incubated for a reaction time of 30 min at 22 C. The concentration of MKNK 2 was adjusted depending of the activity of the enzyme lot and was chosen appropriate to have the assay in the linear range, typical concentrations were in the range of 0.0045 μg/ml. The reaction was stopped by the addition of 5 μΙ of a solution of TR-FRET detection reagents (5 nM streptavidine- XL665 [Cisbio Bioassays, Codolet, France] and 1 nM anti-ribosomal protein S6 (pSer236)- antibody from Invitrogen [# 44921 G] and 1 nM LANCE EU-W1024 labeled ProteinG [Perkin- Elmer, product no. AD0071]) in an aqueous EDTA-solution (100 mM EDTA, 0.1 % (w/v) bovine serum albumin in 50 mM HEPES pH 7.5).

The resulting mixture was incubated for 1 h at 22 C to allow the formation of complex between the phosphorylated biotinylated peptide and the detection reagents. Subsequently the amount of phosphorylated substrate was evaluated by measurement of the resonance energy transfer from the Eu-chelate to the streptavidine-XL665. Therefore, the fluorescence emissions at 620 nm and 665 nm after excitation at 350 nm were measured in a TR-FRET reader, e.g. a Pherastar (BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin- Elmer). The ratio of the emissions at 665 nm and at 622 nm was taken as the measure for the amount of phosphorylated substrate. The data were normalised (enzyme reaction without inhibitor = 0 % inhibition, all other assay components but no enzyme = 100 % inhibition). Usually the test compounds were tested on the same microtiterplate in 1 1 different concentrations in the range of 20 μΜ to 0.1 nM (e.g. 20 μΜ, 5.9 μΜ, 1 .7 μΜ, 0.51 μΜ, 0.15 μΜ, 44 ηΜ, 13 ηΜ, 3.8 ηΜ, 1 .1 ηΜ, 0.33 nM and 0.1 nM, the dilution series prepared separately before the assay on the level of the 10Ofold concentrated solutions in DMSO by serial dilutions, the exact concentrations may vary depending on the pipettor used) in duplicate values for each concentration and ICso values were calculated.

EGFR kinase assay

EGFR inhibitory activity of compounds of the present invention was quantified employing the TR-FRET based EGFR assay as described in the following paragraphs. Epidermal Growth Factor Receptor (EGFR) affinity purified from human carcinoma A431 cells (Sigma-Aldrich, # E3641 ) was used as kinase. As substrate for the kinase reaction the biotinylated peptide biotin-Ahx-AEEEEYFELVAKKK (C-terminus in amid form) was used which can be purchased e.g. form the company Biosynthan GmbH (Berlin-Buch, Germany). For the assay 50 nl_ of a 10Ofold concentrated solution of the test compound in DMSO was pipetted into a black low volume 384well microtiter plate (Greiner Bio-One, Frickenhausen, Germany), 2 μΙ_ of a solution of EGFR in aqueous assay [50 mM Hepes/HCI pH 7.0, 1 mM MgC . 5 mM MnCb. 0.5 mM activated sodium ortho-vanadate, 0.005% (v/v) Tween-20] were added and the mixture was incubated for 15 min at 22°C to allow pre-binding of the test compounds to the enzyme before the start of the kinase reaction. Then the kinase reaction was started by the addition of 3 μΙ_ of a solution of adenosine-tri-phosphate (ATP, 16.7 μΜ => final cone, in the 5 μΐ_ assay volume is 10 μΜ) and substrate (1 .67 μΜ => final cone, in the 5 μΙ_ assay volume is 1 μΜ) in assay buffer and the resulting mixture was incubated for a reaction time of 30 min at 22°C. The concentration of EGFR was adjusted depending of the activity of the enzyme lot and was chosen appropriate to have the assay in the linear range, typical concentration were in the range of 3 U/ml. The reaction was stopped by the addition of 5 μΙ of a solution of HTRF detection reagents (0.1 μΜ streptavidine-XL665 [Cis

Biointernational] and 1 nM PT66-Tb-Chelate, an terbium-chelate labelled anti-phospho- tyrosine antibody from Cis Biointernational [instead of the PT66-Tb-chelate

PT66-Eu-Cryptate from Perkin Elmer can also be used]) in an aqueous EDTA-solution (80 mM EDTA, 0.2 % (w/v) bovine serum albumin in 50 mM HEPES pH 7.5). The resulting mixture was incubated 1 h at 22°C to allow the binding of the biotinylated phosphorylated peptide to the streptavidine-XL665 and the PT66-Eu-Chelate. Subsequently the amount of phosphorylated substrate was evaluated by measurement of the resonance energy transfer from the PT66-Eu-Chelate to the streptavidine-XL665. Therefore, the fluorescence emissions at 620 nm and 665 nm after excitation at 337 nm were measured in a HTRF reader, e.g. a Pherastar (BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin-Elmer). The ratio of the emissions at 665 nm and at 622 nm was taken as the measure for the amount of phosphorylated substrate. The data were normalised (enzyme reaction without inhibitor = 0 % inhibition, all other assay components but no enzyme = 100 % inhibition). Usually the test compounds were tested on the same microtiterplate in 1 1 different concentrations in the range of 20 μΜ to 0.1 n (e.g. 20 μΜ, 5.9 μΜ, 1 .7 μΜ, 0.51 μΜ, 0.15 μΜ, 44 ηΜ, 13 η , 3.8 η , 1 .1 ηΜ, 0.33 nM and 0.1 nM, the dilution series prepared separately before the assay on the level of the 10Ofold concentrated solutions in DMSO by serial dilutions, the exact concentrations may vary depending on the pipettor used) in duplicate values for each concentration and ICso values were calculated.

CDK2/CycE kinase assay

CDK2/CycE inhibitory activity of compounds of the present invention can be quantified employing the CDK2/CycE TR-FRET assay as described in the following paragraphs. Recombinant fusion proteins of GST and human CDK2 and of GST and human CycE, expressed in insect cells (Sf9) and purified by Glutathion-Sepharose affinity chromatography, can be purchased from ProQinase GmbH (Freiburg, Germany). As substrate for the kinase reaction biotinylated peptide biotin-Ttds-YISPLKSPYKISEG (C-terminus in amid form) can be used which can be purchased e.g. from the company JERINI peptide technologies (Berlin, Germany).

For the assay 50 nl_ of a 10Ofold concentrated solution of the test compound in DMSO is pipetted into a black low volume 384well microtiter plate (Greiner Bio-One, Frickenhausen, Germany), 2 μΙ_ of a solution of CDK2/CycE in aqueous assay buffer [50 mM Tris/HCI pH 8.0, 10 mM MgC , 1 .0 mM dithiothreitol, 0.1 mM sodium ortho-vanadate, 0.01 % (v/v) Nonidet-P40 (Sigma)] are added and the mixture is incubated for 15 min at 22°C to allow pre-binding of the test compounds to the enzyme before the start of the kinase reaction. Then the kinase reaction is started by the addition of 3 μΐ_ of a solution of

adenosine-tri-phosphate (ATP, 16.7 μΜ => final cone, in the 5 μΙ_ assay volume is 10 μΜ) and substrate (1 .25 μΜ => final cone, in the 5 μΙ_ assay volume is 0.75 μΜ) in assay buffer and the resulting mixture is incubated for a reaction time of 25 min at 22°C. The

concentration of CDK2/CycE is adjusted depending of the activity of the enzyme lot and is chosen appropriate to have the assay in the linear range, typical concentrations ae in the range of 130 ng/ml. The reaction is stopped by the addition of 5 μΙ_ of a solution of TR-FRET detection reagents (0.2 μΜ streptavidine-XL665 [Cisbio Bioassays, Codolet, France] and 1 n anti-RB(pSer807/pSer81 1 )-antibody from BD Pharmingen [# 558389] and 1 .2 nM LANCE EU-W1024 labeled anti-mouse IgG antibody [Perkin-Elmer, product no. AD0077, as an alternative a Terbium-cryptate-labeled anti-mouse IgG antibody from Cisbio Bioassays can be used]) in an aqueous EDTA-solution (100 mM EDTA, 0.2 % (w/v) bovine serum albumin in 100 mM HEPES/NaOH pH 7.0).

The resulting mixture is incubated 1 h at 22°C to allow the formation of complex between the phosphorylated biotinylated peptide and the detection reagents. Subsequently the amount of phosphorylated substrate is evaluated by measurement of the resonance energy transfer from the Eu-chelate to the streptavidine-XL. Therefore, the fluorescence emissions at 620 nm and 665 nm after excitation at 350 nm is measured in a TR-FRET reader, e.g. a Rubystar (BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin-Elmer). The ratio of the emissions at 665 nm and at 622 nm is taken as the measure for the amount of

phosphorylated substrate. The data are normalised (enzyme reaction without inhibitor = 0% inhibition, all other assay components but no enzyme = 100 % inhibition). Usually the test compounds are tested on the same microtiterplate in 1 1 different concentrations in the range of 20 μΜ to 0.1 nM (20 μΜ, 5.9 μΜ, 1 .7 μΜ. 0.51 μΜ. 0.15 μΜ. 44 nM, 13 nM, 3.8 nM, 1 .1 nM, 0.33 nM and 0.1 nM, the dilution series prepared separately before the assay on the level of the 10Ofold concentrated solutions in DMSO by serial 1 :3.4 dilutions) in duplicate values for each concentration and I CM values are calculated.

PDGFRB kinase assay

PDGFRB inhibitory activity of compounds of the present invention can be quantified employing the PDGFRB HTRF assay as described in the following paragraphs.

As kinase, a GST-His fusion protein containing a C-terminal fragment of human PDGFRB (amino acids 561 - 1 106, expressed in insect cells [SF9] and purified by affinity

chromatography, purchased from Proqinase [Freiburg i.Brsg., Germany] is used. As substrate for the kinase reaction the biotinylated poly-Glu.Tyr (4: 1 ) copolymer (# 61 GT0BLA) from Cis Biointernational (Marcoule, France) is used.

For the assay 50 nl_ of a 10Ofold concentrated solution of the test compound in DMSO is pipetted into a black low volume 384well microtiter plate (Greiner Bio-One, Frickenhausen, Germany), 2 μΙ_ of a solution of PDGFRB in aqueous assay buffer [50 mM HEPES/NaOH pH 7.5, 10 mM MgC , 2.5 mM dithiothreitol, 0.01 % (v/v) Triton-X100 (Sigma)] are added and the mixture was incubated for 15 min at 22°C to allow pre-binding of the test compounds to the enzyme before the start of the kinase reaction. Then the kinase reaction is started by the addition of 3 μΙ_ of a solution of adenosine-tri-phosphate (ATP, 16.7 μΜ => final cone, in the 5 μΙ_ assay volume is 10 μΜ) and substrate (2.27 pg/mi => final cone, in the 5 μΙ_ assay volume is 1 .36 g/ml [~ 30 nM]) in assay buffer and the resulting mixture is incubated for a reaction time of 25 min at 22°C. The concentration of PDGFRB in the assay is adjusted depending of the activity of the enzyme lot and is chosen appropriate to have the assay in the linear range, typical enzyme concentrations are in the range of about 125 pg/μί (final cone, in the 5 μΙ_ assay volume). The reaction is stopped by the addition of 5 μΙ_ of a solution of HTRF detection reagents (200 nM streptavidine-XLent [Cis Biointernational] and 1 .4 nM PT66-Eu-Chelate, an europium-chelate labelled anti-phospho-tyrosine antibody from Perkin Elmer [instead of the PT66-Eu-chelate PT66-Tb-Cryptate from Cis Biointernational can also be used]) in an aqueous EDTA-solution (100 mM EDTA, 0.2 % (w/v) bovine serum albumin in 50 mM HEPES/NaOH pH 7.5).

The resulting mixture is incubated 1 h at 22°C to allow the binding of the biotinylated phosphorylated peptide to the streptavidine-XLent and the PT66-Eu-Chelate. Subsequently the amount of phosphorylated substrate is evaluated by measurement of the resonance energy transfer from the PT66-Eu-Chelate to the streptavidine-XLent. Therefore, the fluorescence emissions at 620 nm and 665 nm after excitation at 350 nm is measured in a HTRF reader, e.g. a Rubystar (BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin-Elmer). The ratio of the emissions at 665 nm and at 622 nm is taken as the measure for the amount of phosphorylated substrate. The data are normalised (enzyme reaction without inhibitor = 0 % inhibition, all other assay components but no enzyme = 100 % inhibition). Normally test compound are tested on the same microtiter plate at 10 different concentrations in the range of 20 μΜ to 1 nM (20 μΜ, 6.7 μΜ, 2.2 μΜ, 0.74 μΜ, 0.25 μΜ, 82 nM, 27 nM, 9.2 nM, 3.1 nM and 1 nM, dilution series prepared before the assay at the level of the 10Ofold cone, stock solutions by serial 1 :3 dilutions) in duplicate values for each concentration and IC 5 o values are calculated.

Fyn kinase assay

C-terminally His6-tagged human recombinant kinase domain of the human T-Fyn expressed in baculovirus infected insect cells (purchased from Invitrogen, P3042) is used as kinase. As substrate for the kinase reaction the biotinylated peptide biotin-KVEKIGEGTYGW

(C-terminus in amid form) is used which can be purchased e.g. form the company

Biosynthan GmbH (Berlin-Buch, Germany).

For the assay 50 nL of a 10Ofold concentrated solution of the test compound in DMSO is pipetted into a black low volume 384well microtiter plate (Greiner Bio-One, Frickenhausen,

Germany), 2 ί of a solution of T-Fyn in ar""~~" "".say buffer [25 mM Tris/HCI pH 7.2, 25 mM MgCb, 2 mM dithiothreitol, 0.1 % (w/v) bovine serum albumin, 0.03% (v/v)

Nonidet-P40 (Sigma)], are added and the mixture is incubated for 15 min at 22°C to allow pre-binding of the test compounds to the enzyme before the start of the kinase reaction. Then the kinase reaction is started by the addition of 3 μΙ_ of a solution of

adenosine-tri-phosphate (ATP, 16.7 μΜ => final cone, in the 5 μΙ_ assay volume is 10 μΜ) and substrate (2 μΜ => final cone, in the 5 μΙ_ assay volume is 1.2 μ ) in assay buffer and the resulting mixture is incubated for a reaction time of 60 min at 22°C. The concentration of Fyn is adjusted depending of the activity of the enzyme lot and is chosen appropriate to have the assay in the linear range, typical concentration was 0.13 nM. The reaction is stopped by the addition of 5 μΙ_ of a solution of HTRF detection reagents (0.2 μΜ streptavidine-XL

[Cisbio Bioassays, Codolet, France) and 0.66 nM PT66-Eu-Chelate, an europium-chelate labelled anti-phospho-tyrosine antibody from Perkin Elmer [instead of the PT66-Eu-chelate PT66-Tb-Cryptate from Cisbio Bioassays can also be used]) in an aqueous EDTA-solution (125 mM EDTA, 0.2 % (w/v) bovine serum albumin in 50 mM HEPES/NaOH pH 7.0). The resulting mixture is incubated 1 h at 22°C to allow the binding of the biotinylated phosphorylated peptide to the streptavidine-XL and the PT66-Eu-Chelate. Subsequently the amount of phosphorylated substrate is evaluated by measurement of the resonance energy transfer from the PT66-Eu-Chelate to the streptavidine-XL. Therefore, the fluorescence emissions at 620 nm and 665 nm after excitation at 350 nm is measured in a HTRF reader, e.g. a Rubystar (BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin-Elmer). The ratio of the emissions at 665 nm and at 622 nm is taken as the measure for the amount of phosphorylated substrate. The data are normalised (enzyme reaction without inhibitor = 0 % inhibition, all other assay components but no enzyme = 100 % inhibition). Normally test compounds are tested on the same microtiter plate at 10 different concentrations in the range of 20 μΜ to 1 nM (20 μΜ, 6.7 μΜ, 2.2 μΜ, 0.74 μΜ, 0.25 μΜ, 82 nM, 27 nM, 9.2 nM, 3.1 nM and 1 nM, dilution series prepared before the assay at the level of the 10Ofold cone, stock solutions by serial 1 :3 dilutions) in duplicate values for each concentration and ICso values are calculated.

Flt4 kinase assay

Flt4 inhibitory activity of compounds of the present invention can be quantified employing the Flt4 TR-FRET assay as described in the following paragraphs.

As kinase, a GST-His fusion protein containing a C-terminal fragment of human Flt4 (amino acids 799 - 1298, expressed in insect cells [SF9] and purified by affinity chromatography, purchased from Proqinase [Freiburg i.Brsg., Germany] is used. As substrate for the kinase reaction the biotinylated peptide Biotin- Ahx-GGEEEEYFELVKKKK (C-terminus in amide form, purchased from Biosyntan, Berlin-Bi"^ < ^--~ a ny) is used. For the assay 50 nl_ of a 10Ofold concentrated solution of the test compound in DMSO was pipetted into a black low volume 384well microtiter plate (Greiner Bio-One, Frickenhausen, Germany), 2 μΙ_ of a solution of Flt4 in aqueous assay buffer [25 mM HEPES pH 7.5, 10 mM MgCb, 2 mM dithiothreitol, 0.01 % (v/v) Triton-X100 (Sigma), 0.5 mM EGTA, and 5 mM β-phospho-glycerol] are added and the mixture is incubated for 15 min at 22°C to allow pre-binding of the test compounds to the enzyme before the start of the kinase reaction. Then the kinase reaction is started by the addition of 3 μΙ_ of a solution of

adenosine-tri-phosphate (ATP, 16.7 μΜ => final cone, in the 5 μΙ_ assay volume is 10 μΜ) and substrate (1.67 μΜ => final cone, in the 5 μΙ_ assay volume is 1 μΜ) in assay buffer and the resulting mixture is incubated for a reaction time of 45 min at 22°C. The concentration of Flt4 in the assay is adjusted depending of the activity of the enzyme lot and was chosen appropriate to have the assay in the linear range, typical enzyme concentrations are in the range of about 120 pg/μΙ. (final cone, in the 5 μΙ_ assay volume). The reaction is stopped by the addition of 5 μΙ_ of a solution of HTRF detection reagents (200 nM streptavidine-XL665 [Cis Biointernational] and 1 nM PT66-Tb-Cryptate, an terbium-cryptate labelled

anti-phospho-tyrosine antibody from Cisbio Bioassays (Codolet, France) in an aqueous EDTA-solution (50 mM EDTA, 0.2 % (w/v) bovine serum albumin in 50 mM HEPES pH 7.5).

The resulting mixture is incubated 1 h at 22°C to allow the binding of the biotinylated phosphorylated peptide to the streptavidine-XL665 and the PT66-Tb-Cryptate. Subsequently the amount of phosphorylated substrate is evaluated by measurement of the resonance energy transfer from the PT66-Tb-Cryptate to the streptavidine-XL665. Therefore, the fluorescence emissions at 620 nm and 665 nm after excitation at 350 nm is measured in a HTRF reader, e.g. a Rubystar (BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin-Elmer). The ratio of the emissions at 665 nm and at 622 nm is taken as the measure for the amount of phosphorylated substrate. The data are normalised (enzyme reaction without inhibitor = 0 % inhibition, all other assay components but no enzyme = 100 % inhibition). Normally test compound are tested on the same microtiter plate at 10 different concentrations in the range of 20 μΜ to 1 nM (20 μΜ, 6.7 μΜ, 2.2 μΜ, 0.74 μΜ, 0.25 μΜ, 82 nM, 27 nM, 9.2 nM, 3.1 nM and 1 nM, dilution series prepared before the assay at the level of the 10Ofold cone, stock solutions by serial 1 :3 dilutions) in duplicate values for each concentration and IC 5 o values are calculated.

TrkA kinase assay

TrkA inhibitory activity of compounds of the present invention can be quantified employing the TrkA HTRF assay as described in the following paragraphs. As kinase, a GST-His fusion protein containing a C-terminal fragment of human TrkA (amino acids 443 - 796, expressed in insect cells rocm purified by affinity chromatography, purchased from Proqinase [Freiburg i.Brsg., Germany] is used. As substrate for the kinase reaction the biotinylated poly-Glu,Tyr (4: 1 ) copolymer (# 61 GT0BLA) from Cis

Biointernational (Marcoule, France) is used.

For the assay 50 nl_ of a 10Ofold concentrated solution of the test compound in DMSO is pipetted into a black low volume 384well microtiter plate (Greiner Bio-One, Frickenhausen, Germany), 2 μΙ_ of a solution of TrkA in aqueous assay buffer [8 mM OPS/HCI pH 7.0, 10 mM MgC , 1 mM dithiothreitol, 0.01 % (v/v) NP-40 (Sigma), 0.2 mM EDTA] are added and the mixture was incubated for 15 min at 22°C to allow pre-binding of the test compounds to the enzyme before the start of the kinase reaction. Then the kinase reaction is started by the addition of 3 μΙ_ of a solution of adenosine-tri-phosphate (ATP, 16.7 μΜ => final cone, in the 5 μΙ_ assay volume is 10 μΜ) and substrate (2.27 μg ml => final cone, in the 5 μΙ_ assay volume is 1 .36 μg/ml [~ 30 nM]) in assay buffer and the resulting mixture is incubated for a reaction time of 60 min at 22°C. The concentration of TrkA in the assay is adjusted depending of the activity of the enzyme lot and is chosen appropriate to have the assay in the linear range, typical enzyme concentrations are in the range of about 20 pg/μί (final cone, in the 5 μΙ_ assay volume). The reaction is stopped by the addition of 5 μΙ_ of a solution of HTRF detection reagents (30 nM streptavidine-XL665 [Cis Biointernational] and 1 .4 nM PT66-Eu-Chelate, an europium-chelate labelled anti-phospho-tyrosine antibody from Perkin Elmer [instead of the PT66-Eu-chelate PT66-Tb-Cryptate from Cis Biointernational can also be used]) in an aqueous EDTA-solution (100 mM EDTA, 0.2 % (w/v) bovine serum albumin in 50 mM HEPES/NaOH pH 7.5).

The resulting mixture is incubated 1 h at 22°C to allow the binding of the biotinylated phosphorylated peptide to the streptavidine-XL665 and the PT66-Eu-Chelate. Subsequently the amount of phosphorylated substrate is evaluated by measurement of the resonance energy transfer from the PT66-Eu-Chelate to the streptavidine-XL665. Therefore, the fluorescence emissions at 620 nm and 665 nm after excitation at 350 nm is measured in a HTRF reader, e.g. a Rubystar (BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin-Elmer). The ratio of the emissions at 665 nm and at 622 nm is taken as the measure for the amount of phosphorylated substrate. The data are normalised (enzyme reaction without inhibitor = 0 % inhibition, all other assay components but no enzyme = 100 % inhibition). Normally test compound are tested on the same microtiter plate at 10 different concentrations in the range of 20 μΜ to 1 nM (20 μΜ, 6.7 μΜ, 2.2 μΜ, 0.74 μΜ, 0.25 μΜ, 82 nM, 27 nM, 9.2 nM, 3.1 nM and 1 nM, dilution series prepared before the assay at the level of the 10Ofold cone, stock solutions by serial 1 :3 dilutions) in duplicate values for each concentration and I CM values are calculated. AlphaScreen SureFire elF4E Ser209 phosphorylation assay

The AlphaScreen SureFire elF4E Ser209 phoshorylation assay can be used to measure the phosphorylation of endogenous elF4E in cellular lysates. The AlphaScreen SureFire technology allows the detection of phosphorylated proteins in cellular lysates. In this assay, sandwich antibody complexes, which are only formed in the presence of the analyte (p-elF4E Ser209), are captured by AlphaScreen donor and acceptor beads, bringing them into close proximity. The excitation of the donor bead provokes the release of singlet oxygen molecules that triggers a cascade of energy transfer in the Acceptor beads, resulting in the emission of light at 520-620nm. Surefire EIF4e Alphascreen in A549 cells with 20% FCS stimulation

For the assay the AlphaScreen SureFire p-elF4E Ser209 10K Assay Kit and the

AlphaScreen ProteinA Kit (for 10K assay points) both from Perkin Elmer are used.

On day one 50.000 A549 cells are plated in a 96-well plate in 100 μΙ_ per well in growth medium (DMEM/Hams' F12 with stable Glutamin, 10%FCS) and incubated at 37°C. After attachment of the cells, medium is changed to starving medium (DMEM, 0.1 % FCS, without Glucose, with Glutamin, supplemented with 5g/L Maltose). On day two, test compounds are serially diluted in 50 μΙ_ starving medium with a final DMSO concentration of 1 % and are added to A549 cells in test plates at a final concentration range from as high 10 μΜ to as low 10 nM depending on the activities of the tested compounds. Treated cells are incubated at 37°C for 2h. 37 ul FCS is added to the wells (=final FCS concentration 20%) for 20 min. Then medium is removed and cells are lysed by adding 50 μΙ_ lysis buffer. Plates are then agitated on a plate shaker for 10 min. After 10 min lysis time, 4μΙ_ of the lysate is transfered to a 384well plate (Proxiplate from Perkin Elmer) and 5μΙ_ Reaction Buffer plus Activation Buffer mix containing AlphaScreen Acceptor beads is added. Plates are sealed with TopSeal-A adhesive film, gently agitated on a plate shaker for 2 hours at room temperature. Afterwards 2μΙ_ Dilution buffer with AlphaScreen Donor beads are added under subdued light and plates are sealed again with TopSeal-A adhesive film and covered with foil. Incubation takes place for further 2h gently agitation at room temperature. Plates are then measured in an EnVision reader (Perkin Elmer) with the AlphaScreen program. Each data point (compound dilution) is measured as triplicate.

Proliferation assays

The tumor cell proliferation assay which can be used to test the compounds of the present invention involves a readout called Cell Titer-Glow ® Luminescent Cell Viability Assay developed by Promega ® (B.A. Cunningham, "A Growing Issue: Cell Proliferation Assays, Modern kits ease quantification of cell growth", The Scientist 2001 , 15(13), 26; S.P. Crouch et al., "The use of ATP bioluminescence a" ~— 'ire of cell proliferation and cytotoxicity", Journal of Immunological Methods 1993. 160. 81-88). that measures inhibition of ceil proliferation. Generation of a luminescent signal corresponds to the amount of ATP present, which is directly proportional to the number of metabolically active (proliferating) cells.

In vitro tumor cell proliferation assay:

Cultivated tumour cells ( OLM-13 (human acute myeloid leukemia cells obtained from DSMZ # ACC 554). JJN-3 (human plasma cell leukemia cells obtained from DS Z # ACC 541 ), Ramos (RA1 ) (human Burkitt's lymphoma cells obtained from ATCC # CRL-159)) are plated at a density of 2,500 cells/well (JJN-3), 3.000 cells/well (MOLM-13), 4.000 cells/well (Ramos (RA1 )), in a 96-well multititer plate (Costa r 3603 black/clear bottom) in 100 pL of their respective growth medium supplemented with 10% fetal calf serum. After 24 hours, the cells of one plate (zero- point plate) are measured for viability. Therefore, 70 pL/well CTG solution (Promega Cell Titer Glo solution (catalog # G755B and G756B)) is added to zero- point plate. The plates are mixed for two minutes on orbital shaker to ensure cell lysis and incubated for ten minutes at room temperature in the dark to stabilize luminescence signal. The samples are read on a VICTOR 3 plate reader. In parallel, serially test compounds are diluted in growth medium, and 50 μΙ_ of 3x dilutions/well are pipetted into the test plates (final concentrations: 0 μΜ, as well as in the range of 0.001-30 μΜ). The final concentration of the solvent dimethyl sulfoxide is 0.3-0.4%. The cells are incubated for 3 days in the presence of test substances. 105 pL/well CTG solution (Promega Cell Titer Glo solution (catalog # G755B and G756B)) is added to the test wells. The plates are mixed for 2 minutes on an orbital shaker to ensure cell lysis and incubated for 10 min at room temperature in the dark to stabilize luminescence signal. The samples are read on a VICTOR 3 plate reader. The change of cell number, in percent, is calculated by normalization of the measured values to the extinction values of the zero-point plate (= 0%) and the extinction of the untreated (0 μιτι) cells (= 100%).

Overview cell lines for proliferation assays

Thus the compounds of the present invention effectively inhibit one or more kinases and are therefore suitable for the treatment or prophylaxis of diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses, particularly in which the uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses is mediated by MKNK, more particularly in which the diseases of uncontrolled cell growth, proliferation and/or survival, inappropriate cellular immune responses, or inappropriate cellular inflammatory responses are haemotological tumours, solid tumours and/or metastases thereof, e.g. leukaemias and myelodysplasia syndrome, malignant lymphomas, head and neck tumours including brain tumours and brain metastases, tumours of the thorax including non-small cell and small cell lung tumours, gastrointestinal tumours, endocrine tumours, mammary and other gynaecological tumours, urological tumours including renal, bladder and prostate tumours, skin tumours, and sarcomas, and/or metastases thereof. In Vitro ΙΙ_-1β (lnterleukin-1 beta, IL-1 b)-induced cytokine secretion of human PBMCs (peripheral biood mononuclear cells)

The effect of the chemical compounds on the induced cytokine secretion of human PBMCs has been investigated. Here, the cytokine secretion has been induced by IL-Ι β which binding to its receptors leads to the activation of the MKNK signaling pathway.

Human PBMCs have been isolated from anti-coagulated human whole blood donated from healthy volunteers by pre-filling Leucosep tubes with ficoll-paque (15 ml, Biochrom, order ID: L61 15) and adding 20 ml whole blood. After centrifugation of the blood at 800g for 15 min at room temperature (RT) plasma including thrombocytes has been discarded. PBMCs were transferred to a new falcon tube, washed with PBS (phosphate-buffered saline) at 250g for 10min at RT and resuspended in complete medium [RPMI 1640, without L-glutamine (PAA, order ID: E15-039), 10% FCS; 50 U/ml Penicillin, 50 pg/ml Streptomycin (PAA, order ID: P1 1 -010) and 1 % L-glutamine (Sigma, order ID. G7513)]. The assay was performed in a 96- well plate at a cell density of 2.5x10 5 cells/well as triplicates. The compounds were serially diluted in DMSO and added to the PBMCs with a final concentration of 0.4% in DMSO, respectively. Treatment of PBMCs with 0.4% DMSO was used as control. After 30 min of incubation, PBMCs were stimulated with 20 ng/ml IL-1 β (R&D, order ID: 201-LB CF) for 24 hours. Cell viability was measured using the CellTiter-Glo Luminescent Assay (Promega, order ID: G7571 ) following the manufacturers protocol. The amount of secreted IL-2, IL-6, IL- 8, IL-10, IL-12p70, GM-CSF, IFN-γ, and TNF-o (tumor necrosis factor-alpha) in the supernatant was determined using the Human Proinflammatory 9-Plex (MSD, order ID: K15007B) according to manufacturer's instruction. The inhibitory activity was determined as the relation to the control in percent. I CM values were calculated using the 4-parameter logistic model.

In Vitro LPS (lipopolysaccharide)-induced cytokine secretion of human PBMCs (peripheral blood mononuclear cells)

The effect of chemical compounds on the induced cytokine secretion of human PBMCs has been investigated. Here, the cytokine secretion has been induced by IL-1 β which binding to its receptor leads to the activation of the MKNK signaling pathway.

Human PBMCs have been isolated from anti-coagulated human whole blood donated from healthy volunteers by pre-filling Leukosep tubes with ficoll-paque (15 ml, Biochrom, order ID: L61 15) and adding 20 ml whole blood. After centrifugation of the whole blood at 800g for 15 min at room temperature (RT) plasma including thrombocytes has been discarded. PBMCs were transferred to a new falcon tube, washed with PBS (phosphate-buffered saline) at 250g for 10 min at RT and resuspended in complete medium [RP I 1640, without L-glutamine (PAA, order ID: E15-039), 10% FCS, 50 U/ml Penicillin, 50 pg/ml Streptomycin (PAA, order ID: P1 1 -010) and 1 % L-glutamine (Sigma, order ID: G7513)]. The assays were performed in a 96-well plate at a cell density of 2.5x10 5 cells/well as triplicates. The compounds were serially diluted in DMSO and added to the PBMCs with a final concentration with 0.4% in DMSO, respectively. Treatment of PBMCs with 0.4% DMSO was used as a control. After 30 min of incubation, PBMCs were stimulated with 100 ng/ml LPS (Lipopolysaccharide, Sigma order ID: L4516) for 24 hours. Cell viability was measured using CellTiter-Glo Luminescent Assay (Promega, order ID: G7571 ) following the manufacturers protocol. The amount of secreted IL-1 β, IL-2, IL-6, IL-8, IL-10, IL-12p70, IFN-γ, and TNF-a in the supernatant was determined using Human Proinflammatory 9-Plex (MSD, order ID. K15007B) or !L-1 β, IL-1 ra, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12p70, IL-13, IL-15, IL-17, eotaxin, basic FGF, G-CSF, GM-CSF, IFN-γ, IP-10, MCP-1 , ΜΙΡ-1 α, ΜΙΡ-1 β, PDGF-BB, RANTES, TNF-a, and VEGF using Bio-Plex Human Group I Assay (Bio-Rad, order ID: M500KCAFOY) according to manufacturer ' s instruction. The inhibitory activity was determined as the relation to the control in percent. IC50 values were calculated.

In Vitro PHA (phytohaemagglutinin)-induced cytokine secretion of human PBMCs (peripheral blood mononuclear cells)

The effect of chemical compounds on the induced cytokine secretion of human PBMCs has been investigated. Here, the cytokine secretion has been induced by IL-1 β which binding to its receptor leads to the activation of the MKNK signaling pathway.

Human PBMCs have been isolated from anti-coagulated human whole blood donated from healthy volunteers by p re-filling Leukosep tubes with ficoll-paque (15 ml, Biochrom, order ID: L61 15) and adding 20 ml whole blood. After centrifugation of the whole blood at 800g for 15 min at room temperature (RT) plasma including thrombocytes has been discarded. PBMCs were transferred to a new falcon tube, washed with PBS (phosphate-buffered saline) at 250g for 10 min at RT and resuspended in complete medium [RPMI 1640, without L-glutamine (PAA, order ID: E15-039), 10% FCS, 50 U/ml Penicillin, 50 pg/ml Streptomycin (PAA, order ID: P1 1 -010) and 1 % L-glutamine (Sigma, order ID: G7513)]. The assays were performed in a 96-well plate at a cell density of 2.5x10 5 cells/well as triplicates. The compounds were serially diluted in DMSO and added to the PBMCs with a final concentration with 0.4% in DMSO, respectively. Treatment of PBMCs with 0.4% DMSO was used as a control. After 30 min of incubation, PBMCs were stimulated with 10 mg/ml PHA (Phytohaemagglutinine, Sigma, order ID: L4144) for 24 hours. Cell viability was measured using CellTiter-Glo Luminescent Assay (Promega, order ID: G7571 ) following the manufacturers protocol. The amount of secreted pro-inflammatory cytokines (e.g. IL-1 β, IL-4, IL-6, IL-10, IL-17A, IL-17F, IL-21 , IL-22, IL-23, IL-25, IL-31 , IL-33, CD40L, TNF-ct, IFN-γ) in the supernatant was determined using Bio-Plex Human Assay (Bio-Rad, order ID: 171AA001 ) according to manufacturer's instruction. The inhibitory activity was determined as the relation to the control in percent. I CM values were calculated.

In vitro - 3T3-L1 differentiation into adipocyte-like cells

The effect of chemical compounds on the differentiation of pre-adipocytes into adipocytes has been investigated. Here, 3T3-L1 fibroblasts were differentiated into adipocyte-like cells. Preadipocytes (3T3-L1 ) were grown to 2 days post-confluence in DMEM supplemented with 10% BCS (day 0) and the medium was changed to DMEM supplemented with 10% FBS, insulin (167 nM), dexamethasone (0.5 μΜ), isobutylmethylxanthine (IBMX) (0.5 mM) and rosiglitazone (2 μΜ) and test compounds. After 48 h, the medium replaced by DMEM supplemented with 10% (v/v) FBS and 167 nM insulin and different concentrations of test compound. This maintenance medium was changed every 48 h always containing different concentrations of the test compound. Cells underwent the differentiation program up to 9 days. Gene expression of MNK1 , MNK2 as well as of adipocyte differentiation marker such as PPARy, C/EBPa, SREBPI c, GLUT4, CD36, FAS, cytosolic phospholipase A2a was performed. The relative mRNA expression of each gene in compound treated cells was compared to the relative mRNA expression in differentiated but untreated control cells.

Oil Red O staining was used to qualify and quantify intracellular triglyceride levels. For quantification, cells were washed extensively with water to remove unbound dye, and 1 ml isopropanol was added to the stained cells. Photometric evaluation was applied.

Lipid incorporation was also measured by triglyceride assay and lipolysis assay. Lipolysis describes the hydrolysis of triglycerides into glycerol and free fatty acids. Lipolysis is induced by the synthetic catecholamine, Isoproterenol and glycerol serves as a measure for the incorporated fat during differentiation into adipocytes. For the lipolysis assay, 3T3-L1 cells were differentiated, as described above, for 9 days. The lipolysis assay was performed according to the manufacturer's instructions (e.g. Abeam Lipolysis assay kit, ab185433). The amount of glycerol released was measured using colorimetric intensity.

In Vivo Models

In Vivo CFA (Complete Freund's Adjuvant)-induced inflammatory pain in rats

The complete Freund's adjuvant (CFA)-induced inflammatory pain model can be used to evaluate the effect of the chemical compounds on pain.

On day 0, rats (Sprague-Dawley, 10 animals/group) receive either vehicle or the chemical compound before injection of CFA followed by daily treatment until day 7. To inject 100 μΙ of 100% CFA (Sigma, order ID: F5881 ) subcutaneously into the plantar surface of the left hind paw rats are anesthetized with 2.5 - 5% isoflurane in oxygen. Mechanical allodynia is assessed using von Frey filaments according to the "up-down" method described by Chaplan et al (1994) on day 7. Here, calibrated monofilaments (von Frey filaments) are applied to the plantar surface of the rat hind paw for a period of 4-6 seconds, or until a nocifensive paw withdrawal occurres.

In vivo Imiquimod-induced Psoriasis model in mice

Chemical compounds were tested for their effects in an Imiquimod (IMQ)-induced psoriasis model in mice.

Induction of psoriasis was performed on animals by daily administration of Imiquimod after shaving and depilation of the animals at Day -1. 3.5 mg of IMQ (equivalent to 70 mg of Aldara® creme, Meda AB) was topically administered from Day 1 to Day 7 (seven days) in the morning for groups to be induced. Sham group was applied with paraffin oil. The surface of application was about 4 cm 2 on the back of the animals which corresponds to a rectangle of about 1.5 cm to 2.5 cm on the back of the animal. The cream was smoothly massaged on the back for 5 seconds. After this time, the internal face of the right ear was massaged with remaining cream from finger glove. Then, the back was massaged again for 5 seconds and afterwards the internal face of the left ear was massaged with the remaining cream from finger gloves. Animals were weighed every second days (Day 1 prior induction, Day 3, Day 5 and Day 7). Due to body weight loss observed with this model, 0.4 ml of sterile saline was injected intraperitoneally (ip) at Day 3 and Day 4. Oral (po) administration of test compounds started at Day 1 not more than 1 hour after IMQ treatment. The test compounds was administered once daily from Day 1 to Day 7. In each treatment group n = 10 animals were included with a Sham (no IMQ) group, a negative control group (IMQ plus vehicle), two groups treated with IMQ and different dosages of the test compound, and one group with IMQ treated animals receiving the reference (betamethasone) compound. Development of the disease was determined using a clinical scoring system. Every day, the following clinical scores were recorded from back skin

Both ears and back skin thickness was measured daily using digital sliding caliper (Horex Digital Caliper, Helios Preisser, Germany). At Day 4, 4 hours after morning treatment, blood was collected from the mandibular vein under isofluran anesthesia into heparin sodium tubes (0.5 ml Eppendorf tubes). After blood collection, 0.4 ml of sterile saline was injected ip to the animals. At Day 7 about 500 μΙ blood were collected after intercardiac puncture from isofluran-anesthetized in to heparin sodium tubes. Blood was centrifuged (3500 g for 10 min at 5°C. Plasma was separated and stored at -80°C for further analyses (determination of cytokine levels). At Day 7 animals were euthanized, and back skin calibrated digital punctures were taken. Skin samples from the back and both ears were kept in formalin and placed in 70% ethanol 48 hours after collection. Histopathological evaluation of back and ear skin was done after hematoxylin and eosin (HE) staining by an experienced Pathologist.

In vivo OG33-35-induced, chronic EAE (Experimental Autoimmune Encephalomyelitis)- model in mice

The effects of chemical compounds were evaluated in an experimental autoimmune encephalomyelitis (EAE) model in C57BI/6 mice.

On Day 0 all mice, except the animals from healthy control group were given a subcutaneous (sc) injection of 0.1 ml on two sites on the back of 200 pg MOG35-55 (myelin oligodendrocyte glycoprotein) emulsified in Complete Freund's Adjuvant supplemented with mycobacterium tuberculosis. At this same time point, mice were injected with 200 ng of Pertussis Toxin (PT) dissolved in 0.1 mi PBS (phosphate buffered saline); the PT injection was repeated on Day 2. A group of ten animals served as naive, vehicle-dosed control group. The other groups underwent induction of EAE and received test compound treatment. Mice were weighed daily. Symptoms of EAE were assessed daily, starting on Day 4 and continuing through study end. All remaining mice were sacrificed at the completion of the study (Day 30). Blood was collected for preparation of serum. Animals were then perfused with formalin, and the spinal cord was collected and stored in 10% formalin for subsequent histopathology analysis. Slides were evaluated by a broad certified veterinary pathologist and were assessed for inflammation and demyelination on a 0-5 scoring scale. Terminal serum samples were analyzed via multiplex analyses for levels of TNF-a, IL-6. IL-12, IL-23, ΙΙ_-1 β, IL-17, and IFNy.

In vivo Adjuvant-induced arthritis in rats

The effects of chemical compounds on the adjuvant-induced arthritis in rats were

investigated.

At day 0 male Lewis rats (100 to 125 g body weight, Charles River Laboratories, Germany) were treated at the tail subcutaneously (sc) with 100 μΙ of complete Freund's Adjuvant (CFA) solution [M. tuberculosis H37Ra (Difco Lab, cat. No. 231 141 )] diluted in Incomplete Freund's Adjuvant (Difco Lab, cat.no: 263910). Animals were randomized with n=8 animals per treatment group. As controls a healthy and a disease group treated with vehicle only were included in the studies. Treatment with test compounds was done orally (po) with either one or more dosages using an appropriate vehicle which allowed sufficient exposure of the animals with the test compound. In a preventive treatment setting, treatment start was at day 0 and continued to day 20, the end of the study. Observation of the disease induction as well as the treatment effects of test compound was determined by the RA disease activity score, starting at day 0 and then three times per week. The Score defines the extent of joint inflammation from 0 to 4 including erythema with swelling of the joint (0 = no; 1 = slight: 2 = moderate, 3 = important, 4 = very important). The disease score was determined for both hind paws and added to one value. As an additional parameter for joint swelling the paw volume was determined using a plethysometer (IITC Life Science Inc., USA). A second parameter determined during the study was the grip strength using an automated grip strength test meter (IITC Life Science Inc., USA) as a measure for hyperalgesia. At the end of the study (day 20) synovial fluid from joints, biopsies of kneejoint and blood serum were obtained and used for determination of proinflammatory cytokines [Meso Scale Discovery (MSD), Proinflammatory Panel 1 ; cat no: K15059D), and the c-reactive protein (CRP) (BD Biosciences, cat no: 55825).

Synovial fluid is isolated by rinsing the inflamed joints with 150 μΙ sterile sodium acetate solution. Biopsies of kneejoints were homogenized with a cryo mill (Retsch GmbH, Germany) at -196°C. 200 mg of the powder were used for analysis of cytokines suspended in 0.5 ml RP I1640 medium. Statistical analyses of obtained results were done with one way Anova (ANOVA; Analysis of variance) and the comparison to the control group via multiple reference analysis (Dunnett-test).

In vivo - Fertility model in mice

Chemical compound were tested for their effects on fertility in a DBA/2J-CBA/J mouse model. Female CBA/J mice bred with mal DBA/2J display a higher abortion rate.

Male DBA/2J mice were bred with female CBA/J mice and the vaginal plugs in individual mated female mice were examined daily to determine potential pregnancy. At Day 1 of pregnancy one or two groups of successfully mated female CBA J mice were treated orally (po) with one or two different dosages of the test compound once daily, whereas the control group was given the vehicle only. In each treatment group n = 12 animals were included. At Day 10 to Day 14 of pregnancy the animals were sacrificed, the uteri removed and the implantation sites were documented. The abortion sites were identified by their small size and necrotic, hemorrhagic appearance, compared with normal embryos and placentas. The percentage of abortions was calculated as the ratio of resorption sites to total implantation sites. Uteri were shock frozen and after homogenization cytokine levels were determined using mouse Bio-Plex Assays (M0009 DPD, M6000007NY, LJ00000163).

In vivo CCL4-induced liver fibrosis in mice

The effect of chemical compounds was tested in a tetrachlormethan (CCL4)-induced mouse model of liver fibrosis.

Eight week old 90 male C57BI/6 mice (Charles River) were randomly divided into three groups (Group 1 = untreated control, Group 2= vehicle treated control, Group 3 = treatment with test compound) with n = 30 in each group. For induction of liver fibrosis animals were treated three times per week (Monday, Wednesday, Friday) with 50 μΙ of CCL4/olive oil suspension (CCL4 + olive oil, 1 +9 v/v) intraperitoneal^ (ip) over the whole study time. CCL4 is the most commonly used inducer of a toxically-induced liver fibrosis in animal models (Starkel et al., 201 1 ). Once daily per os (po) treatment of group 2 with the vehicle and of group 3 with test compound suspension started with the first day and was done over the complete study duration. Two weeks after study start fifty percent of animals (of each group) were euthanized and after two additional weeks, the remaining animals were euthanized. After finalization of the study, the liver of each animal as collected and fixed in 4% formaldehyde and paraffin embedded for further histopathological analyses. For

determination of severity of liver fibrosis liver slices were stained with pikro-sirius red (Waldeck, Germany) to visualize the collagen content in the tissue. A Carl Zeiss microscope (Axio) connected to a PC was used to scan the pikro-sirius red stained liver sections to make images of these. The sections were scanned at a magnification of 20x and a light intensity of 4.8V. The images were then formatted into jpg and the red -stained area quantified by using the ImageJ Software (National Institue of Health, USA). The results are expressed as % sirius-red per liver area.

Chemical compounds of the invention are tested for their effects in following in models

In vivo - Letrozole-induced Polycystic Ovary Syndrome in rats

Chemical compounds are tested for their effects in a Polycystic Ovary Syndrome (PCOS) model in rats.

Han Wistar rats are randomly divided into 3 groups with n=8 animals per group. Rats in the control group receive vehicle only once daily per os (po), whereas rats of the other two groups were all administered letrozole at a concentration of 1 mg/kg body weight dissolved in 0.5 % carboxymethylcellulose (CMC) once daily for consecutive 28 days. Animals of the third group additionally receive the test compound once daily po in 20% HPBCD (HP beta cyclodextrin) for consecutive 28 days. Vaginal smears are performed, and the rat weights are recorded daily. At day 27 animals are fasted and an oral glucose tolerance test will be performed. Rats are euthanized, ovaries are removed and weighed. One ovary of each rat is fixed in formaldehyde for histological examination, whereas the other ovary is stored at -80°C for mRNA and protein analyses. Fat tissue and liver are also removed and from each tissue one aliquot is fixed in formaldehyde and the other aliquot is shock frozen for further mRNA analyses.

Vaginal Smears: the stage of the estrous cycle is determined by microscopic analysis of the predominant cell type in the daily vaginal smears.

Ovarian morphology: sections from the ovarian tissue are taken from the part of the ovary with largest diameter, stained with hematoxylin and eosin.

In vivo - diet induced obesity in mice

Effects of chemical compound on diet-induced obesity are tested in C57BI/6 mice receiving a high fat diet.

Male C57BI/6 (n = 45) mice at an age of 16 weeks are randomly divided into 5 groups.

Animals fo groups 1 to 4 are fed with 60% high fat diet for 10 weeks. Animals of group 5 (n = 10) are fed with chow diet for the same time. Mice of group 1 are additionally treated orally (BID) with vehicle only. Animals of group 2 (n=15) receive test compound (BID) orally. Mice of group 3 (n=10) receive pioglitazone via oral administration (QD). Compound and vehicle treatment is performed over the complete study duration. Animals of groups 4 (n=10) and 5 (n=10) do not receive any treatment. Body weight will be measured daily during the 8 weeks treatment period. At days 26 and 48 of treatment overnight fasted mice will undergo an oral glucose tolerance test (OGTT). Mice will be weighed and blood (15 μΙ/EDTA) will be collected from the tail tip two hours after compound treatment; 30 min prior glucose load to measure HBA1 c, blood glucose, and plasma insulin and HOMAR-IR will then be calculated.

HOMAR-IR index = [(mM glucose X pU/rnl insulin)/22.5]

For OGTT blood glucose will be measured from tail tip at -30, 0, 15, 30, 60, 90, and 120 min after oral glucose load (1.5 g/kg body weight). Blood (20 μΙ/EDTA) is also collected at 15 and 30 min after glucose load to measure plasma insulin. Blood glucose levels are used to calculate glucose area under the curve. Mice recover from the OGTT over 2 days.

At days 28 of treatment blood (15 μΙ/EDTA) is collected 2 hours after compound treatment from the tail tip to measure HBA1 c, blood glucose and plasma insulin in fed conditions and HOMAR-IR will then be calculated.

At day 51 of treatment 4 h fasted mice will then undergo an Insulin Tolerance Test (ITT) with insulin (0.5 U/kg body weight) injected intraperitoneally. Blood glucose will be then measured at time 0, 15, 30, 60, 90, and 120 minutes after insulin injection. At day 53 of treatment the incorporated body fat is determined by Echo- I. At day 56 of treatment (end of the study) blood is taken for determination of blood glucose, leptin, triglycerides, total cholesterol, and free fatty acids. In vivo pharmacokinetics in rats

For in vivo pharmacokinetic experiments test compounds were administered to male Wistar rats intravenously at doses of 0.3 to 1 mg/kg and intragastrai at doses of 0.5 to 10 mg/kg formulated as solutions using solubilizers such as PEG400 in well-tolerated amounts. For pharmacokinetics after intravenous administration test compounds were given as i.v. bolus and blood samples were taken at 2 min, 8 min, 15 min, 30 min, 45 min, 1 h, 2 h, 4 h, 6 h, 8 h and 24 h after dosing. Depending on the expected half-life additional samples were taken at later time points (e.g. 48 h, 72 h). For pharmacokinetics after intragastrai administration test compounds were given intragastrai to fasted rats and blood samples were taken at 5 min, 15 min, 30 min, 45 min, 1 h, 2 h, 4 h, 6 h, 8 h and 24 h after dosing.

Depending on the expected half-life additional samples were taken at later time points (e.g. 48 h, 72 h). Blood was collected into Lithium-Heparintubes ( onovetten ® , Sarstedt) and centrifuged for 15 min at 3000 rpm. An aliquot of 100 μΙ_ from the supernatant (plasma) was taken and precipitated by addition of 400 μΙ_ cold acetonitril and frozen at -20 °C over night. Samples were subsequently thawed and centrifuged at 3000 rpm, 4°C for 20 minutes.

Aliquots of the supernatants were taken for analytical testing using an Agilent 1200 HPLC- system with LCMS/MS detection. PK parameters were calculated by non-compartmental analysis using a PK calculation software. PK parameters derived from concentration-time profiles after i.v.: CLplasma: Total plasma clearance of test compound (in L/kg/h); CLblood: Total blood clearance of test compound: CLplasma*Cp/Cb (in L/kg/h) with Cp/Cb being the ratio of concentrations in plasma and blood. PK parameters calculated from concentration time profiles after i.g.: Cmax: Maximal plasma concentration (in mg/L); Cmaxnorm: Cmax divided by the administered dose (in kg/L); Tmax: Time point at which Cmax was observed (in h). Parameters calculated from both, i.v. and i.g. concentration-time profiles: AUCnorm: Area under the concentration-time curve from t=0h to infinity (extrapolated) divided by the administered dose (in kg*h/L);

AUC(0-tlast)norm: Area under the concentration-time curve from t=0h to the last time point for which plasma concentrations could be measured divided by the administered dose (in kg*h/L); t1 /2: terminal half-life (in h); F: oral bioavailability: AUCnorm after intragastrai administration divided by AUCnorm after intravenous administration (in %).