Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SUBSTITUTED PYRAZOLYL-BASED CARBOXAMIDE AND UREA DERIVATIVES BEARING A PHENYL MOIETY SUBSTITUTED WITH AN N-CONTAINING GROUP AS VANILLOID RECEPTOR LIGANDS
Document Type and Number:
WIPO Patent Application WO/2013/068462
Kind Code:
A1
Abstract:
The invention relates to substituted pyrazolyl-based carboxamide and urea derivatives of formula (R) bearing a phenyl moiety substituted with an N-containing group as vanilloid receptor ligands, to pharmaceutical compositions containing these compounds and also to these compounds for use in the treatment and/or prophylaxis of pain and further diseases and/or disorders.

Inventors:
FRANK ROBERT (DE)
CHRISTOPH THOMAS (DE)
DAMANN NILS (DE)
LESCH BERNHARD (DE)
BAHRENBERG GREGOR (DE)
SAUNDERS DEREK JOHN (DE)
STOCKHAUSEN HANNELORE (DE)
KIM YONG-SOO (KR)
KIM MYEONG-SEOP (KR)
LEE JEEWOO (KR)
Application Number:
PCT/EP2012/072141
Publication Date:
May 16, 2013
Filing Date:
November 08, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
GRUENENTHAL GMBH (DE)
International Classes:
C07D231/12; A61K31/415; A61P29/00
Domestic Patent References:
WO2010127856A12010-11-11
WO2010127855A12010-11-11
WO2010127856A12010-11-11
Other References:
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
BENNETT, G.J.; XIE, Y.K.: "A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man", PAIN, vol. 33, no. 1, 1988, pages 87 - 107, XP024378950, DOI: doi:10.1016/0304-3959(88)90209-6
KIM, S.H.; CHUNG, J.M.: "An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat", PAIN, vol. 50, no. 3, 1992, pages 355 - 363, XP024378108, DOI: doi:10.1016/0304-3959(92)90041-9
D'AMOUR; SMITH, J. PHARM. EXP. THER., vol. 72, 1941, pages 74 - 79
D. DUBUISSON ET AL., PAIN, vol. 4, 1977, pages 161 - 174
J. MARCH: "Advanced Organic Chemistry", 2007, WILEY & SONS
F. A. CAREY; R. J. SUNDBERG: "Advanced Organic Chemistry", 2007, SPRINGER
"Compendium of Organic Synthetic Methods", WILEY & SONS
CHENG; PRUSOFF, BIOCHEM. PHARMACOL., vol. 22, 1973, pages 3099 - 3108
Attorney, Agent or Firm:
BÜLLE, Jan et al. (Köln, DE)
Download PDF:
Claims:
Patent claims:

1 . A substituted compound of general formula (R),

(R).

in which

R 101 103

, R and R are independently of one another selected from the group consisting of H, F, CI, Br, CFH2, CF2H, CF3, CN, CH2-OH, CH2CH2-OH, CH2- OCH3, CH2CH2-OCH3, OCFH2, OCF2H, OCF3, OH, NH2, a d_4 alkyl, an 0-d_4 alkyl, a NH-Ci-4 alkyl, and a N(Ci_4 alkyl)2, wherein the Ci-4 alkyl is in each case unsubstituted,

R2 represents CF3, an unsubstituted Ci-4 alkyl or an unsubstituted C3.6 cycloalkyl,

R7 and R9 are independently of one another selected from the group consisting of H, F, CI, Br, CFH2, CF2H, CF3, CN, OH, OCF3, a d-4 alkyl, and an O-C1-4 alkyl, wherein the Ci-4 alkyl is in each case unsubstituted,

A denotes N, CH or C(CH3), q denotes 0, 1 or 2,

R 2 represents H or a Ci-4 alkyl, which is unsubstituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents selected from the group consisting of F, CI, Br, OH, =0 and OCH3,

R 3 represents a H, S(=0)2-NH2, a Ci-4 alkyl or a S(=0)2-C1.4 alkyl, wherein the Ci-4 alkyl is in each case unsubstituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents selected from the group consisting of F, CI, Br, OH, =0 and OCH3, or - with the proviso that q is≠ 0 - R 2 and R113 together with the nitrogen atom connecting them form a 3 to 6 membered heterocyclyl, which is unsusbtituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents independently of one another selected from the group consisting of F, CI, Br, CN, CF3, CH3, CH2CH3, CH(CH3)2, tert.-butyl, cyclopropyl, OH, =0, OCH3, OCF3, NH2, NH(CH3) and N(CH3)2, optionally in the form of a single stereoisomer or a mixture of stereoisomers, in the form of the free compound and/or a physiologically acceptable salt thereof.

2. The compound according to claim 1 , characterized in that R2 represents CF3, tert.-butyl or cyclopropyl.

3. The compound according to claim 1 or 2, characterized in that

R 0 , R 02 and R 03 are independently of one another selected from the group consisting of H, F, CI, Br, CFH2, CF2H, CF3, CN, CH2-OH, CH2-OCH3, OCF3, OH, CH3, CH2CH3, CH(CH3)2, 0-CH3, 0-CH2CH3, NH2, NH(CH3), and N(CH3)2.

4. The compound according to any of the preceding claims, characterized in that A denotes N,

R 2 represents H or a d-4 alkyl, which is unsubstituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents selected from the group consisting of F, CI, Br, OH, =0 and OCH3,

R 3 represents a H, S(=0)2-NH2, a d-4 alkyl or a S(=0)2-d-4 alkyl, wherein the d- alkyl is in each case unsubstituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents selected from the group consisting of F, CI, Br, OH, =0 and OCH3, or - with the proviso that q is≠ 0 -

R 2 and R113 together with the nitrogen atom connecting them form a 3 to 6 membered heterocyclyl, which is unsusbtituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents independently of one another selected from the group consisting of F, CI, Br, CN, CF3, CH3, CH2CH3, CH(CH3)2, tert.-butyl, cyclopropyl, OH, =0, OCH3, OCF3, NH2, NH(CH3) and N(CH3)2, or

A denotes CH or C(CH3),

R 2 represents H or a Ci-4 alkyl, which is unsubstituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents selected from the group consisting of F, CI, Br, OH, =0 and OCH3,

R 3 represents a H, S(=0)2-NH2, a Ci-4 alkyl, which is unsubstituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents selected from the group consisting of F, CI, Br, OH, =0 and OCH3, or - with the proviso that q is≠ 0 -

R 2 and R 3 together with the nitrogen atom connecting them form a 3 to 6 membered heterocyclyl, which is unsusbtituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents independently of one another selected from the group consisting of F, CI, Br, CN, CF3, CH3, CH2CH3, CH(CH3)2, tert.-butyl, cyclopropyl, OH, =0, OCH3, OCF3, NH2, NH(CH3) and N(CH3)2.

5. The compound according to any of the preceding claims, characterized in that q denotes 0, 1 or 2,

A denotes N,

R 0 is selected from the group consisting of H, F, CI, Br, CFH2, CF2H, CF3, CN, CH2-OH, CH2-OCH3, OCF3, OH, CH3, CH2CH3, CH(CH3)2, 0-CH3, 0-CH2CH3, NH2, NH(CH3) and N(CH3)2, and

R 02 and R 03 are independently of one another selected from the group consisting of H, F, CI, Br, CFH2, CF2H, CF3, CN, CH2-OH, CH2-OCH3, OCF3, OH, CH3, CH2CH3, CH(CH3)2, 0-CH3, 0-CH2CH3, NH2, NH(CH3) and N(CH3)2, or q denotes 1 or 2,

A denotes CH or C(CH3),

R 0 is selected from the group consisting of H, F, Br, CFH2, CF2H, CF3, CN, CH2- OH, CH2-OCH3, OCF3, OH, CH3, CH2CH3, CH(CH3)2, 0-CH3, 0-CH2CH3, NH2, NH(CH3) and N(CH3)2, and

103

R and R are independently of one another selected from the group consisting of H, F, CI, Br, CFH2, CF2H, CF3, CN, CH2-OH, CH2-OCH3, OCF3, OH, CH3, CH2CH3, CH(CH3)2, 0-CH3, 0-CH2CH3, NH2, NH(CH3) and N(CH3)2.

6. The compound according to any of the preceding claims, characterized in that at least one of R 0 , R 02 and R 03 is≠ H.

7. The compound according to any of the preceding claims, characterized in that

R7 and R9 are independently of one another selected from the group consisting of H, F, CI, Br, CF3, CN, OH, OCF3, CH3, CH2CH3, CH(CH3)2, 0-CH3, and 0-CH2CH3.

8. The compound according to any of the preceding claims, characterized in that at least one of R7 and R9 is≠ H.

9. The compound according to any of claims 1 -3 and 5-8, characterized in that the part structure (RS1 )

q

(RS1 ) represents the part structure (PR1 )

(PR1 ),

wherein

R 4 represents NH2 or an unsubstituted d-4 alkyl.

10. The compound according to any of claims 1 -3 and 5-9, characterized in that the part structure (RS1 )

q

(RS1 ) represents the part structure (PR1 )

(PR1 ),

wherein

Rm represents NH2, CH3 or CH2CH3.

1 1 . The compound according to any of the preceding claims, characterized in that A denotes N or C(CH3).

12. The compound according to any one of claims 1 -5, 7, 9 and 10, characterized in that A denotes N and R 0 is selected from the group consisting of H, F, CI, Br, CFH2, CF2H, CF3, CN, CH2-OH, CH2-OCH3, OCF3, OH, CH3, CH2CH3, CH(CH3)2, 0-CH3, 0-CH2CH3, NH2, NH(CH3) and N(CH3)2, and

103

R and R are independently of one another selected from the group consisting of H, F, CI, Br, CFH2, CF2H, CF3, CN, CH2-OH, CH2-OCH3, OCF3, OH, CH3, CH2CH3, CH(CH3)2, 0-CH3, 0-CH2CH3, NH2, NH(CH3) and N(CH3)2, or

A denotes CH or C(CH3) and

R 0 is selected from the group consisting of H, F, Br, CFH2, CF2H, CF3, CN, CH2- OH, CH2-OCH3, OCF3, OH, CH3, CH2CH3, CH(CH3)2, 0-CH3, 0-CH2CH3, NH2, NH(CH3) and N(CH3)2, and

103

R 02 and R are independently of one another selected from the group consisting of H, F, CI, Br, CFH2, CF2H, CF3, CN, CH2-OH, CH2-OCH3, OCF3, OH, CH3, CH2CH3, CH(CH3)2, 0-CH3, 0-CH2CH3, NH2, NH(CH3) and N(CH3)2;

R2 represents CF3, tert.-butyl or cyclopropyl,

R7 and R9 are independently of one another selected from the group consisting of H, F, CI, Br, CF3, CN, OH, OCF3, CH3, CH2CH3, CH(CH3)2, 0-CH3, and 0-CH2CH3, the part structure (RS1 )

q

(RS1 ) represents the part structure (

(PR1 ), wherein

Rm represents NH2, CH3 or CH2CH3.

13. The compound according to any one of the preceding claims, selected from the group consisting of

B1 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-

4- (methanesulfonamido-methyl)-phenyl]-propionamide;

B2 N-[[2-(3-Chloro-4-fluoro-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-

[3-fluoro-4-(methanesulfonamido-methyl)-phenyl]-propionamide;

B3 N-[[5-tert-Butyl-2-(3-chloro-4-fluoro-phenyl)-2H-pyrazol-3-yl]-methyl]-2-[3- fluoro-4-(methanesulfonamido-methyl)-phenyl]-propionamide;

B4 N-[[2-(3-Chlorophenyl)-5-cyclopropyl-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B5 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-[4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B6 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B7 2-[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-N-[[2-(3-fluorophenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-propionamide;

B8 2-[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-N-[[5-(trifluoromethyl)-2-

[3-(trifluoromethyl)phenyl]-2H-pyrazol-3-yl]-methyl]-propionamide;

B9 N-[[5-tert-Butyl-2-(3-fluorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B10 N-[[5-tert-Butyl-2-(3,4-difluoro-phenyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B11 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

(methanesulfonamido-methyl)-3-methoxy-phenyl]-urea;

B12 1 -[[2-(3-Chlorophenyl)-5-(trif luoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

(methanesulfonamido-methyl)-3-methoxy-phenyl]-urea;

B13 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[3-chloro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B14 1 -[[2-(3-Chlorophenyl)-5-(trif luoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

(methylamino-methyl)-phenyl]-urea;

B15 2-[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-N-[[2-(3-methoxyphenyl)-

5- (trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-propionamide; B16 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[4-

[[(ethylsulfonyl)amino]-methyl]-3-fluoro-phenyl]-propionamide;

B17 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-[4-

[[(ethylsulfonyl)amino]-methyl]-3-fluoro-phenyl]-propionamide;

B18 N-[[2-(4-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-

4-(methanesulfonamido-methyl)-phenyl]-propionamide;

B19 N-[[2-(3,4-Difluoro-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-[3- fluoro-4-(methanesulfonamido-methyl)-phenyl]-propionamide;

B20 N-[[5-tert-Butyl-2-(4-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B21 N-[[5-tert-Butyl-2-(4-fluorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B22 2-[3-Chloro-4-(methanesulfonamido-methyl)-phenyl]-N-[[2-(3-chlorophenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-propionamide;

B23 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-

4-(methanesulfonamido-methyl)-phenyl]-acetamide;

B24 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

(ethylamino-methyl)-3-fluoro-phenyl]-urea;

B25 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[4-(ethylamino- methyl)-3-fluoro-phenyl]-urea;

B26 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-acetamide;

B27 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[3,5- difluoro-4-(methanesulfonamido-methyl)-phenyl]-urea;

B28 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

(methanesulfonamido-methyl)-phenyl]-urea;

B29 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-

4-[(methyl-methylsulfonyl-amino)-methyl]-phenyl]-propionamide;

B30 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-4-

[(methyl-methylsulfonyl-amino)-methyl]-phenyl]-propionamide;

B31 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B32 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[3,5-difluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B33 N-[[4-[[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl- carbamoyl]amino]-2-fluoro-phenyl]-methyl]-acetamide; B34 N-[[4-[[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl- carbamoyl]amino]-2-fluoro-phenyl]-methyl]-acetamide;

B35 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B36 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B37 2-[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-N-[[2-(3-fluorophenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-acetamide;

B38 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

(dimethylaminomethyl)-3-fluoro-phenyl]-urea;

B39 1 -[4-(Aminomethyl)-3-fluoro-phenyl]-3-[[2-(3-chlorophenyl)-5-(trifluoromethyl)-

2H-pyrazol-3-yl]-methyl]-urea;

B40 1 -[4-(Aminomethyl)-3-fluoro-phenyl]-3-[[5-tert-butyl-2-(3-chlorophenyl)-2H- pyrazol-3-yl]-methyl]-urea;

B41 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[3-fluoro-

4-[(sulfamoylamino)-methyl]-phenyl]-urea;

B42 2-[4-(Aminomethyl)-3-fluoro-phenyl]-N-[[2-(3-chlorophenyl)-5-(trifluoromethyl)-

2H-pyrazol-3-yl]-methyl]-propionamide;

B43 N-[[5-tert-Butyl-2-[3-(trifluoromethyl)phenyl]-2H-pyrazol-3-yl]-methyl]-2-[3- fluoro-4-(methanesulfonamido-methyl)-phenyl]-propionamide;

B44 1 -[[2-(3-Fluorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

(methanesulfonamido-methyl)-phenyl]-urea;

B45 1 -[[5-tert-Butyl-2-(3-fluorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

(methanesulfonamido-methyl)-phenyl]-urea;

B46 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B47 2-[4-(Aminomethyl)-3-fluoro-phenyl]-N-[[5-tert-butyl-2-(3-chlorophenyl)-2H- pyrazol-3-yl]-methyl]-propionamide;

B48 1 -[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-3-[[2-(3-fluorophenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea;

B49 1 -[[2-(3,4-Difluoro-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[3- fluoro-4-(methanesulfonamido-methyl)-phenyl]-urea;

B50 1 -[[5-tert-Butyl-2-(3-fluorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B51 1 -[[5-tert-Butyl-2-(3-chloro-4-fluoro-phenyl)-2H-pyrazol-3-yl]-methyl]-3-[3- fluoro-4-(methanesulfonamido-methyl)-phenyl]-urea; B52 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[3-fluoro-

4- (methanesulfonamido-methyl)-phenyl]-urea;

B53 1 -[[5-tert-Butyl-2-(4-fluorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B54 1 -[[5-tert-Butyl-2-[3-(trifluoromethyl)phenyl]-2H-pyrazol-3-yl]-methyl]-3-[3- fluoro-4-(methanesulfonamido-methyl)-phenyl]-urea;

B55 1 -[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-3-[[5-(trifluoromethyl)-2-

[3-(trifluoromethyl)phenyl]-2H-pyrazol-3-yl]-methyl]-urea;

B56 1 -[[5-tert-Butyl-2-(3-fluorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B57 1 -[[2-(3,4-Difluoro-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B58 1 -[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-3-[[2-(3-methoxyphenyl)-

5- (trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea;

B59 1 -[[5-tert-Butyl-2-(3-fluorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B60 1 -[[2-(3-Fluorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[3-fluoro-

4-[(sulfamoylamino)-methyl]-phenyl]-urea;

B61 1 -[[2-(3-Chlorophenyl)-5-cyclopropyl-2H-pyrazol-3-yl]-methyl]-3-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B62 1 -[[2-(3,4-Difluoro-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[3- fluoro-4-[(sulfamoylamino)-methyl]-phenyl]-urea;

B63 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-4-

(methylamino-methyl)-phenyl]-propionamide;

B64 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[4-

(dimethylaminomethyl)-3-fluoro-phenyl]-propionamide;

B65 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-[4-

(dimethylaminomethyl)-3-fluoro-phenyl]-propionamide;

B66 2-[4-(Acetylamino-methyl)-3-fluoro-phenyl]-N-[[2-(3-chlorophenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-propionamide;

B67 2-[4-(Acetylamino-methyl)-3-fluoro-phenyl]-N-[[5-tert-butyl-2-(3-chlorophenyl)-

2H-pyrazol-3-yl]-methyl]-propionamide;

B68 2-[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-N-[[2-(m-tolyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-propionamide;

B69 1 -[[5-tert-Butyl-2-(3,4-difluoro-phenyl)-2H-pyrazol-3-yl]-methyl]-3-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-urea; B70 1 -[[2-(3-Chloro-4-fluoro-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-

[3-fluoro-4-[(sulfamoylamino)-methyl]-phenyl]-urea;

B71 1 -[3-Fluoro-4-[(sulfamoylamino)-methyl]-phenyl]-3-[[2-(3-methoxyphenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea;

B72 1 -[[5-tert-Butyl-2-(3,4-difluoro-phenyl)-2H-pyrazol-3-yl]-methyl]-3-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B73 1 -[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-3-[[2-(3-isopropyl-phenyl)-

5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea;

B74 1 -[[5-tert-Butyl-2-(3,4-difluoro-phenyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B75 1 -[[2-(3-Chlorophenyl)-5-cyclopropyl-2H-pyrazol-3-yl]-methyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B76 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-[4-

(methanesulfonamido-methyl)-3-methoxy-phenyl]-propionamide;

B77 1 -[[2-(3-Fluorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B78 1 -[[2-(3-Chloro-4-fluoro-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-

[4-[(sulfamoylamino)-methyl]-phenyl]-urea;

B79 1 -[[2-(3-lsopropyl-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B80 1 -[3-Fluoro-4-[(sulfamoylamino)-methyl]-phenyl]-3-[[2-(3-isopropyl-phenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea;

B81 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

(methanesulfonamido-methyl)-phenyl]-urea;

B82 2-[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-N-[[2-(3-isopropyl- phenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-propionamide;

B83 1 -[[2-(3-lsopropyl-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

(methanesulfonamido-methyl)-phenyl]-urea;

B84 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

[[(ethylsulfonyl)amino]-methyl]-phenyl]-urea;

B85 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

[[(ethylsulfonyl)amino]-methyl]-phenyl]-urea;

B86 1 -[4-(Methanesulfonamido-methyl)-3-methoxy-phenyl]-3-[[2-(m-tolyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea;

B87 1 -[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-3-[[2-(m-tolyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea; B88 N-[[5-tert-Butyl-2-(m-tolyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B89 1 -[4-(Methanesulfonamido-methyl)-phenyl]-3-[[2-(m-tolyl)-5-(trifluoromethyl)^

2H-pyrazol-3-yl]-methyl]-urea;

B90 1 -[[5-tert-Butyl-2-(m-tolyl)-2H-pyrazol-3-yl]-methyl]-3-[4-(methanesulfonamido- methyl)-phenyl]-urea;

B91 1 -[4-[[(Ethylsulfonyl)amino]-methyl]-phenyl]-3-[[2-(m-tolyl)-5-(trifluoromethy^

2H-pyrazol-3-yl]-methyl]-urea;

B92 1 -[[5-tert-Butyl-2-(m-tolyl)-2H-pyrazol-3-yl]-methyl]-3-[4-[[(ethylsulfonyl)amino]- methyl]-phenyl]-urea;

B93 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-propionamide;

B94 2-[3-Fluoro-4-[(sulfamoylamino)-methyl]-phenyl]-N-[[2-(3-isopropyl-phenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-propionamide;

B95 N-[[2-(3-Chlorophenyl)-5-cyclopropyl-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-propionamide;

B96 N-[[5-tert-Butyl-2-(3-chloro-4-fluoro-phenyl)-2H-pyrazol-3-yl]-methyl]-2-[3- fluoro-4-[(sulfamoylamino)-methyl]-phenyl]-propionamide;

B97 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-[3-fluoro-

4-[(sulfamoylamino)-methyl]-phenyl]-propionamide;

B98 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[4-

(methanesulfonamido-methyl)-3-methoxy-phenyl]-propionamide;

B99 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

(pyrrolidin-1 -yl-methyl)-phenyl]-urea;

B100 1 -[[2-(3-Chlorophenyl)-5-(trif luoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[3-f luoro-

4-(piperidin-1 -yl-methyl)-phenyl]-urea;

B101 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-3-[3-fluoro-4-

(pyrrolidin-1 -yl-methyl)-phenyl]-urea;

B102 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-[4-

[(sulfamoylamino)-methyl]-phenyl]-propionamide;

B103 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[4-

[(sulfamoylamino)-methyl]-phenyl]-propionamide;

B104 N-[[2-(3-Fluorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-[4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B105 1 -[[2-(3-Fluorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-3-[4-

(methanesulfonamido-methyl)-3-methoxy-phenyl]-urea; B106 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-2-[4- (methanesulfonamido-methyl)-3-methyl-phenyl]-propionamide; and

B107 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]-2-[4- (methanesulfonamido-methyl)-3-methyl-phenyl]-propionamide; optionally in the form of a single stereoisomer or a mixture of stereoisomers, in the form of the free compound and/or a physiologically acceptable salt thereof.

14. A pharmaceutical composition comprising at least one substituted compound

according to any one of claims 1 to 13.

15. A substituted compound according to any one of claims 1 to 13 for use in the treatment and/or prophylaxis of one or more diseases and/or disorders selected from the group consisting of pain, preferably of pain selected from the group consisting of acute pain, chronic pain, neuropathic pain, visceral pain and joint pain; hyperalgesia; allodynia; causalgia; migraine; depression; nervous affection; axonal injuries; neurodegenerative diseases, preferably selected from the group consisting of multiple sclerosis, Alzheimer's disease, Parkinson's disease and Huntington's disease; cognitive dysfunctions, preferably cognitive deficiency states, particularly preferably memory disorders; epilepsy; respiratory diseases, preferably selected from the group consisting of asthma, bronchitis and pulmonary inflammation; coughs; urinary incontinence; overactive bladder (OAB); disorders and/or injuries of the gastrointestinal tract; duodenal ulcers; gastric ulcers; irritable bowel syndrome; strokes; eye irritations; skin irritations; neurotic skin diseases; allergic skin diseases; psoriasis; vitiligo; herpes simplex; inflammations, preferably inflammations of the intestine, the eyes, the bladder, the skin or the nasal mucous membrane; diarrhoea; pruritus; osteoporosis; arthritis; osteoarthritis; rheumatic diseases; eating disorders, preferably selected from the group consisting of bulimia, cachexia, anorexia and obesity; medication dependency; misuse of medication; withdrawal symptoms in medication dependency; development of tolerance to medication, preferably to natural or synthetic opioids; drug dependency; misuse of drugs; withdrawal symptoms in drug dependency; alcohol dependency; misuse of alcohol and withdrawal symptoms in alcohol dependency; for diuresis; for antinatriuresis; for influencing the cardiovascular system; for increasing vigilance; for the treatment of wounds and/or burns; for the treatment of severed nerves; for increasing libido; for modulating movement activity; for anxiolysis; for local anaesthesia and/or for inhibiting undesirable side effects, preferably selected from the group consisting of hyperthermia, hypertension and bronchoconstriction, triggered by the administration of vanilloid receptor 1 (VR1 /TRPV1 receptor) agonists, preferably selected from the group consisting of capsaicin, resiniferatoxin, olvanil, arvanil, SDZ- 249665, SDZ-249482, nuvanil and capsavanil.

Description:
Substituted pyrazolyl-based carboxamide and urea derivatives bearing a phenyl moiety substituted with an N-containing group as vanilloid receptor ligands

FIELD OF THE INVENTION

The invention relates to substituted pyrazolyl-based carboxamide and urea derivatives bearing a phenyl moiety substituted with an N-containing group as vanilloid receptor ligands, to pharmaceutical compositions containing these compounds and also to these compounds for use in the treatment and/or prophylaxis of pain and further diseases and/or disorders.

BACKGROUND OF THE INVENTION

The treatment of pain, in particular of neuropathic pain, is very important in medicine. There is a worldwide demand for effective pain therapies. The urgent need for action for a patient- focused and target-oriented treatment of chronic and non-chronic states of pain, this being understood to mean the successful and satisfactory treatment of pain for the patient, is also documented in the large number of scientific studies which have recently appeared in the field of applied analgesics or basic research on nociception.

The subtype 1 vanilloid receptor (VR1/TRPV1 ), which is often also referred to as the capsaicin receptor, is a suitable starting point for the treatment of pain, in particular of pain selected from the group consisting of acute pain, chronic pain, neuropathic pain and visceral pain. This receptor is stimulated inter alia by vanilloids such as capsaicin, heat and protons and plays a central role in the formation of pain. In addition, it is important for a large number of further physiological and pathophysiological processes and is a suitable target for the therapy of a large number of further disorders such as, for example, migraine, depression, neurodegenerative diseases, cognitive disorders, states of anxiety, epilepsy, coughs, diarrhoea, pruritus, inflammations, disorders of the cardiovascular system, eating disorders, medication dependency, misuse of medication and urinary incontinence.

Compounds which have an affinity for the subtype 1 vanilloid receptor (VR1/TRPV1 ) are e.g. known from WO 2010/127855-A2 and WO 2010/127856-A2. There is a demand for further compounds having comparable or better properties, not only with regard to affinity to vanilloid receptors 1 (VR1/TRPV1 receptors) per se (potency, efficacy).

Thus, it may be advantageous to improve the metabolic stability, the solubility in aqueous media or the permeability of the compounds. These factors can have a beneficial effect on oral bioavailability or can alter the PK/PD (pharmacokinetic/pharmacodynamic) profile; this can lead to a more beneficial period of effectiveness, for example.

A weak or non-existent interaction with transporter molecules, which are involved in the ingestion and the excretion of pharmaceutical compositions, is also to be regarded as an indication of improved bioavailability and at most low interactions of pharmaceutical compositions. Furthermore, the interactions with the enzymes involved in the decomposition and the excretion of pharmaceutical compositions should also be as low as possible, as such test results also suggest that at most low interactions or no interactions at all, of pharmaceutical compositions are to be expected.

It was therefore an object of the invention to provide novel compounds, preferably having advantages over the prior-art compounds. The compounds should be suitable in particular as pharmacological active ingredients in pharmaceutical compositions, preferably in pharmaceutical compositions for the treatment and/or prophylaxis of disorders or diseases which are at least partially mediated by vanilloid receptors 1 (VR1/TRPV1 receptors).

This object is achieved by the subject matter described herein.

It has surprisingly been found that the substituted compounds of general formula (R), as given below, display outstanding affinity to the subtype 1 vanilloid receptor (VR1 /TRPV1 receptor) and are therefore particularly suitable for the prophylaxis and/or treatment of disorders or diseases which are at least partially mediated by vanilloid receptors 1 (VR1/TRPV1 ).

Particularly suitable are substituted compounds of general formula (R), as given below, that in addition to their activity with regard to the VR1 -receptor show one or more additional advantageous properties, for example, suitable potency, suitable efficacy, no increase in body temperature and/or heat pain threshold; appropriate solubility in biologically relevant media such as aqueous media, in particular in aqueous media at a physiologically acceptable pH value, such as in buffer systems, for instance in phosphate buffer systems; suitable metabolic stability and diversity (e.g. sufficient stability towards the oxidative capabilities of hepatic enzymes such as cytochrome P450 (CYP) enzymes and sufficient diversity with regard to the metabolic elimination via these enzymes); and the like.

The present invention therefore relates to a substituted compound of general formula (R),

(R).

in which

R 101 , R 02 and R 03 are independently of one another selected from the group consisting of H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 CH 2 -OH, CH 2 -OCH 3 , CH 2 CH 2 - OCH 3 , OCFH 2 , OCF 2 H, OCF3, OH, NH 2 , a d- 4 alkyl, an O-C 1 -4 alkyl, a NH-C 1 -4 alkyl, and a N(Ci- 4 alkyl) 2 , wherein the Ci -4 alkyl is in each case unsubstituted,

R 2 represents CF 3 , an unsubstituted Ci -4 alkyl or an unsubstituted C 3 - 6 cycloalkyl,

R 7 and R 9 are independently of one another selected from the group consisting of H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, OH, OCF 3 , a d_ 4 alkyl, and an 0-d_ 4 alkyl, wherein the d_ 4 alkyl is in each case unsubstituted,

A denotes N, CH or C(CH 3 ), q denotes 0, 1 or 2,

R 2 represents H or a Ci -4 alkyl, which is unsubstituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents selected from the group consisting of F, CI, Br, OH, =0 and OCH 3 ,

R 3 represents a H, S(=0) 2 -NH 2 , a Ci- 4 alkyl or a alkyl, wherein the Ci- 4 alkyl is in each case unsubstituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents selected from the group consisting of F, CI, Br, OH, =0 and OCH 3 , or - with the proviso that q is≠ 0 -

R 2 and R 3 together with the nitrogen atom connecting them form a 3 to 6 membered heterocyclyl, which is unsusbtituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents independently of one another selected from the group consisting of F, CI, Br, CN, CF 3 , CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , tert.-butyl, cyclopropyl, OH, =0, OCH 3 , OCF 3 , NH 2 , NH(CH 3 ) and N(CH 3 ) 2 , optionally in the form of a single stereoisomer or a mixture of stereoisomers, in the form of the free compound and/or a physiologically acceptable salt and/or a physiologically acceptable solvate thereof.

DETAILED DESCRIPTION

The term "single stereoisomer" preferably means in the sense of the present invention an individual enantiomer or diastereomer. The term "mixture of stereoisomers" means in the sense of this invention the racemate and mixtures of enantiomers and/or diastereomers in any mixing ratio.

The term "physiologically acceptable salt" preferably comprises in the sense of this invention a salt of at least one compound according to the present invention and at least one physiologically acceptable acid or base.

A physiologically acceptable salt of at least one compound according to the present invention and at least one physiologically acceptable acid preferably refers in the sense of this invention to a salt of at least one compound according to the present invention with at least one inorganic or organic acid which is physiologically acceptable - in particular when used in human beings and/or other mammals. Examples of physiologically acceptable acids are: hydrochloric acid, hydrobromic acid, sulphuric acid, methanesulphonic acid, p- toluenesulphonic acid, carbonic acid, formic acid, acetic acid, oxalic acid, succinic acid, tartaric acid, mandelic acid, fumaric acid, maleic acid, lactic acid, citric acid, glutamic acid, saccharic acid, monomethylsebacic acid, 5-oxoproline, hexane-1 -sulphonic acid, nicotinic acid, 2, 3 or 4-aminobenzoic acid, 2,4,6-trimethylbenzoic acid, oc-lipoic acid, acetyl glycine, hippuric acid, phosphoric acid, aspartic acid. Citric acid and hydrochloric acid are particularly preferred. Hydrochloride salts and citrate salts are therefore particularly preferred salts.

A physiologically acceptable salt of at least one compound according to the present invention and at least one physiologically acceptable base preferably refers in the sense of this invention to a salt of at least one compound according to the present invention as an anion with at least one preferably inorganic cation, which is physiologically acceptable - in particular when used in human beings and/or other mammals. Particularly preferred are the salts of the alkali and alkaline earth metals but also ammonium salts [NH X R 4 . X ] + , in which x = 0, 1 , 2, 3 or 4 and R represents a branched or unbranched Ci -4 alkyl residue, in particular (mono-) or (di)sodium, (mono-) or (di)potassium, magnesium or calcium salts.

The terms "alkyl" and "Ci -4 alkyl" preferably comprise in the sense of this invention acyclic saturated aliphatic hydrocarbon residues, which can be respectively branched or unbranched and can be unsubstituted or can be mono- or polysubstituted, e.g. mono-, di- or trisubstituted, and which contain 1 to 4, i.e. 1 , 2, 3 or 4, carbon atoms, i.e. Ci -4 aliphatic residues, i.e. Ci -4 alkanyls. Preferred Ci -4 alkanyl residues are selected from the group consisting of methyl, ethyl, n-propyl, 2-propyl, n-butyl, isobutyl, sec-butyl, and tert.-butyl.

In relation to the terms "alkyl" and "Ci -4 alkyl", the term "monosubstituted" or "polysubstituted" such as di- or tri-substituted refers in the sense of this invention, with respect to the corresponding residues or groups, to the single substitution or multiple substitution, e.g. disubstitution or trisubstitution, of one or more hydrogen atoms each independently of one another by at least one substituent. The term "polysubstituted" such as di- or tri-substituted with respect to polysubstituted residues and groups such as di- or tri-substituted residues and groups includes the polysubstitution of these residues and groups either on different or on the same atoms, for example trisubstituted on the same carbon atom, as in the case of CF 3 or CH 2 CF 3 or at various points, as in the case of CH(OH)-CH 2 CH 2 -CHCI 2 . The multiple substitution can be carried out using the same or using different substituents.

The terms "cycloalkyl" and "C 3 - 6 cycloalkyl" preferably mean for the purposes of this invention cyclic aliphatic (cycloaliphatic) hydrocarbons containing 3, 4, 5, or 6 carbon atoms, i.e. C 3 - 6 - cycloaliphatic residues, wherein the hydrocarbons are saturated, and which can be unsubstituted or can be mono- or polysubstituted, e.g. mono-, di- or trisubstituted. The cycloalkyl can be bound to the respective superordinate general structure via any desired and possible ring member of the cycloalkyl residue. Preferably, cycloalkyl is selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl, more preferably from the group consisting of cyclopropyl and cyclobutyl. A particularly preferred cycloalkyl is cyclopropyl.

The terms "heterocyclyl" and "3 to 6 membered heterocyclyl" preferably comprise in the sense of this invention aliphatic saturated heterocycloalkyls having 3 to 6, i.e. 3, 4, 5, or 6, ring members, i.e. a 3 to 6 membered heterocyclyl, in which at least one, if appropriate also two or three carbon atoms are replaced by a heteroatom or a heteroatom group each selected independently of one another from the group consisting of O, S, S(=0), S(=0) 2 , N, NH and N(Ci -8 alkyl), wherein the ring members can be unsubstituted or can be mono- or polysubstituted, e.g. mono-, di- or trisubstituted. Heterocyclyls are thus heterocycloaliphatic residues. A heterocyclyl according of the present invention is formed from radicals R 2 and R 3 together with the nitrogen atom connecting them, i.e. contains at least one N as a ring member. Heterocyclyl residues from the group comprising azetidinyl, aziridinyl, dithiolanyl, dihydropyrrolyl, dihydropyridinyl, imidazolidinyl, isoxazolidinyl, morpholinyl, pyrrolidinyl, piperazinyl, 4-methylpiperazinyl, piperidinyl, pyrazolidinyl, tetrahydropyrrolyl, tetrahydropyridinyl, thiazolidinyl and thiomorpholinyl are preferred.

In relation to the terms "cycloalkyl, "C 3 . 6 cycloalkyl", "heterocyclyl" and "3 to 6 membered heterocyclyl", the term "monosubstituted" or "polysubstituted" such as di- or tri-substituted refers in the sense of this invention, with respect to the corresponding residues or groups, to the single substitution or multiple substitution, e.g. disubstitution or trisubstitution, of one or more hydrogen atoms each independently of one another by at least one substituent. The term "polysubstituted" such as di- or tri-substituted with respect to polysubstituted residues and groups such as di- or tri-substituted residues and groups includes the polysubstitution of these residues and groups either on different or on the same atoms, for example disubstituted on the same carbon atom, as in the case of 1 ,1 -difluorocyclohexyl, or at various points, as in the case of 1 -chloro-3-fluorocyclohexyl. The multiple substitution can be carried out using the same or using different substituents.

Within the scope of the present invention, the symbol

used in the formulae denotes a link of a corresponding residue to the respective superordinate general structure.

In a preferred embodiment of the compound according to the present invention

R 101 , R 02 and R 03 are independently of one another selected from the group consisting of H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 .

Preferably, R 101 , R 02 and R 03 are independently of one another selected from the group consisting of H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , O- CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 .

More preferably,

R 101 , R 02 and R 03 are independently of one another selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OCH 3 , OCF 3 , CH 3 , 0-CH 3 , 0-CH 2 CH 3 and N(CH 3 ) 2 .

Even more preferably,

R 101 , R 02 and R 03 are independently of one another selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , OCF 3 , CH 3 , 0-CH 3 , and 0-CH 2 CH 3 .

Still more preferably,

R 101 , R 02 and R 03 are independently of one another selected from the group consisting of H, F, CI, CF 3 , OCF 3 , CH 3 and 0-CH 3 .

Particularly,

R 101 , R 02 and R 03 are independently of one another selected from the group consisting of H, F, CI, CF 3 and 0-CH 3 .

Even more particularly preferred

R 101 , R 02 and R 03 are independently of one another selected from the group consisting of H, F, CI and 0-CH 3 .

In a preferred embodiment of the compound according to the present invention at least one of R 101 , R 02 and R 03 is≠ H.

In another preferred embodiment of the compound according to the present invention one or two of R 101 , R 02 and R 03 , preferably R 02 and/or R 03 , denote(s) H. In another preferred embodiment of the compound according to the present invention one of R 0 , R 02 and R 03 represents H, preferably R 03 represents H.

In another preferred embodiment of the compound according to the present invention

R 0 and R 02 are independently of one another selected from the group consisting of

H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 , and R 03 represents H.

Preferably,

R 0 and R 02 are independently of one another selected from the group consisting of

H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 , more preferably are independently of one another selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OCH 3 , OCF 3 , CH 3 , 0-CH 3 , O- CH 2 CH 3 and N(CH 3 ) 2 , even more preferably are independently of one another selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , OCF 3 , CH 3 , 0-CH 3 , and 0-CH 2 CH 3 , still more preferably are independently of one another selected from the group consisting of H, F, CI, CF 3 , OCF 3 , CH 3 and 0-CH 3 , in particular are independently of one another selected from the group consisting of H, F, CI, CF 3 and 0-CH 3 , even more particularly preferred are independently of one another selected from the group consisting of H, F, CI, and 0-CH 3 , and R 03 represents H.

In yet another preferred embodiment of the compound according to the present invention

R 0 is selected from the group consisting of F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 - OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 , and both R 02 and R 03 represents H.

Preferably,

R 0 is selected from the group consisting of F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 - OCH 3 , OCF 3 , OH, CH 3 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 , more preferably is selected from the group consisting of F, CI, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OCH 3 , OCF 3 , CH 3 , O- CH 3 , 0-CH 2 CH 3 and N(CH 3 ) 2 , even more preferably is selected from the group consisting of F, CI, CFH 2 , CF 2 H, CF 3 , OCF 3 , CH 3 , 0-CH 3 , and 0-CH 2 CH 3 , still more preferably is selected from the group consisting of F, CI, CF 3 , OCF 3 , CH 3 and 0-CH 3 , in particular is selected from the group consisting of F, CI, CF 3 and 0-CH 3 , even more particularly preferred is selected from the group consisting of F, CI, and 0-CH 3 , and both R 02 and R 03 represents H.

In still another preferred embodiment of the compound according to the present invention

R 02 is selected from the group consisting of F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 - OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 , and both R 0 and R 03 represents H.

Preferably,

R 02 is selected from the group consisting of F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 - OCH 3 , OCF 3 , OH, CH 3 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 , more preferably is selected from the group consisting of F, CI, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OCH 3 , OCF 3 , CH 3 , O- CH 3 , 0-CH 2 CH 3 and N(CH 3 ) 2 , even more preferably is selected from the group consisting of F, CI, CFH 2 , CF 2 H, CF 3 , OCF 3 , CH 3 , 0-CH 3 , and 0-CH 2 CH 3 , still more preferably is selected from the group consisting of F, CI, CF 3 , OCF 3 , CH 3 and 0-CH 3 , in particular is selected from the group consisting of F, CI, CF 3 and 0-CH 3 , even more particularly preferred is selected from the group consisting of F, CI, and 0-CH 3 , and both R 0 and R 03 represents H.

In yet a further preferred embodiment of the compound according to the present invention

R 0 is selected from the group consisting of F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 - OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 ,

R 02 is selected from the group consisting of H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 , and R 03 represents H. Preferably,

R 0 is selected from the group consisting of F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 - OCH 3 , OCF3, OH, CH 3 , O-CH3, 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 , more preferably is selected from the group consisting of F, CI, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OCH 3 , OCF 3 , CH 3 , O- CH 3 , 0-CH 2 CH 3 and N(CH 3 ) 2 , even more preferably is selected from the group consisting of F, CI, CFH 2 , CF 2 H, CF 3 , OCF 3 , CH 3 , 0-CH 3 , and 0-CH 2 CH 3 , still more preferably is selected from the group consisting of F, CI, CF 3 , OCF 3 , CH 3 and 0-CH 3 , in particular is selected from the group consisting of F, CI, CF 3 and 0-CH 3 , even more particularly preferred is selected from the group consisting of F, CI, and 0-CH 3 ,

R 02 is selected from the group consisting of H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 , more preferably is selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , CN, CH 2 - OCH 3 , OCF 3 , CH 3 , 0-CH 3 , 0-CH 2 CH 3 and N(CH 3 ) 2 , even more preferably is selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , OCF 3 , CH 3 , 0-CH 3 , and 0-CH 2 CH 3 , still more preferably is selected from the group consisting of H, F, CI, CF 3 , OCF 3 , CH 3 and O- CH 3 , in particular is selected from the group consisting of H, F, CI, CF 3 and 0-CH 3 , even more particularly preferred is selected from the group consisting of H, F, CI, and 0-CH 3 , and R 03 represents H.

In yet another further preferred embodiment of the compound according to the present invention

R 0 is selected from the group consisting of F, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 - OCH 3 , OCF 3 , OH, CH 3 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 , more preferably is selected from the group consisting of F, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OCH 3 , OCF 3 , CH 3 , O- CH 3 , 0-CH 2 CH 3 and N(CH 3 ) 2 , even more preferably is selected from the group consisting of F, CFH 2 , CF 2 H, CF 3 , OCF 3 , CH 3 , 0-CH 3 , and 0-CH 2 CH 3 , still more preferably is selected from the group consisting of F, CF 3 , OCF 3 , CH 3 and 0-CH 3 , in particular is selected from the group consisting of F, CF 3 and 0-CH 3 , even more particularly preferred is selected from the group consisting of F and 0-CH 3 , and both R 02 and R 03 represents H. In still another further preferred embodiment of the compound according to the present invention

R 0 is selected from the group consisting of F, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 - OCH 3 , OCF3, OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 ,

R 02 is selected from the group consisting of H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 , and R 03 represents H. Preferably,

R 0 is selected from the group consisting of F, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 - OCH 3 , OCF 3 , OH, CH 3 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 , more preferably is selected from the group consisting of F, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OCH 3 , OCF 3 , CH 3 , O- CH 3 , 0-CH 2 CH 3 and N(CH 3 ) 2 , even more preferably is selected from the group consisting of F, CFH 2 , CF 2 H, CF 3 , OCF 3 , CH 3 , 0-CH 3 , and 0-CH 2 CH 3 , still more preferably is selected from the group consisting of F, CF 3 , OCF 3 , CH 3 and 0-CH 3 , in particular is selected from the group consisting of F, CF 3 and 0-CH 3 , even more particularly preferred is selected from the group consisting of F and 0-CH 3 ,

R 02 is selected from the group consisting of H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ), and N(CH 3 ) 2 , more preferably is selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , CN, CH 2 - OCH 3 , OCF 3 , CH 3 , 0-CH 3 , 0-CH 2 CH 3 and N(CH 3 ) 2 , even more preferably is selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , OCF 3 , CH 3 , 0-CH 3 , and 0-CH 2 CH 3 , still more preferably is selected from the group consisting of H, F, CI, CF 3 , OCF 3 , CH 3 and O- CH 3 , in particular is selected from the group consisting of H, F, CI, CF 3 and 0-CH 3 , even more particularly preferred is selected from the group consisting of H, F, CI, and 0-CH 3 ,

In another particularly preferred embodiment according to the present invention the part structure (RS2)

(RS2), is selected from the group consisting of

in particular when q denotes 0, 1 or 2, and A denotes N. Even more particularly preferred, the part structure (RS2)

(RS2), is selected from the group consisting of

in particular when q denotes 0, 1 or 2, and A denotes N.

Most preferred, the part structure (RS2)

(RS2), is selected from the group consisting of

in particular when q denotes 0, 1 or 2, and A denotes N, preferably is selected from the group consisting of in particular when q denotes 0, 1 or 2, and A denotes N.

In another particularly preferred embodiment according to the present invention the part structure (RS2)

is selected from the group consisting of

in particular when q denotes 1 or 2, and A denotes CH or C(CH 3 ).

Even more particularly preferred, the part structure (RS2)

(RS2),

in particular when q denotes 1 or 2, and A denotes CH or C(CH 3 ), preferably is selected from the group consisting of in particular when q denotes 1 or 2, and A denotes CH or C(CH 3 ).

In another preferred embodiment of the compound according to the present invention

R 2 represents CF 3 , methyl, ethyl, n-propyl, 2-propyl, n-butyl, iso-butyl, sec-butyl, tert.- butyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.

Preferably,

R 2 represents CF 3 , 2-propyl, n-butyl, iso-butyl, sec-butyl, tert.-butyl, cyclopropyl, or cyclobutyl.

More preferably,

R 2 represents CF 3 , tert.-butyl or cyclopropyl.

In a particularly preferred embodiment of the compound according to the present invention R 2 represents CF 3 .

In another particularly preferred embodiment of the compound according to the present invention R 2 represents tert.-butyl.

In another particularly preferred embodiment of the compound according to the present invention R 2 represents cyclopropyl.

In a further preferred embodiment of the compound according to the present invention

R 7 and R 9 are independently of one another selected from the group consisting of H, F, CI, Br, CF 3 , CN, OH, OCF 3 , CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , and 0-CH 2 CH 3 .

Preferably, R 7 and R 9 are independently of one another selected from the group consisting of H, F, CI, CF 3 , CN, OH, OCF 3 , CH 3 , 0-CH 3 , and 0-CH 2 CH 3 .

More preferably,

R 7 and R 9 are independently of one another selected from the group consisting of H, F, CI, CF 3 , O-CH3, and 0-CH 2 CH 3 .

Even more preferably,

R 7 and R 9 are independently of one another selected from the group consisting of H, F, CI, and 0-CH 3 , still more preferably are independently of one another selected from the group consisting of H, F and CI.

In yet a further preferred embodiment of the compound according to the present invention at least one of R 7 and R 9 is≠ H.

In a further preferred embodiment of the compound according to the present invention R 9 denotes H.

In yet another preferred embodiment of the compound according to the present invention

R 7 is selected from the group consisting of F, CI, Br, CF 3 , CN, OH, OCF 3 , CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , and 0-CH 2 CH 3 , preferably is selected from the group consisting of F, CI, CF 3 , CN, OH, OCF 3 , CH 3 , 0-CH 3 , and 0-CH 2 CH 3 , more preferably is selected from the group consisting of F, CI, CF 3 , 0-CH 3 , and 0-CH 2 CH 3 , even more preferably is selected from the group consisting of F, CI, and 0-CH 3 , still more preferably is selected from the group consisting of F and CI, and R 9 represents H.

In another preferred embodiment of the compound according to the present invention A denotes N or C(CH 3 ).

In a particularly preferred embodiment of the compound according to the present invention A denotes N. In another particularly preferred embodiment of the compound according to the present invention A denotes C(CH 3 ).

In another preferred embodiment of the compound according to the present invention q denotes 1 or 2, preferably 1 .

In a further preferred embodiment of the compound according to the present invention A denotes N, and

R 2 represents H or a Ci -4 alkyl, which is unsubstituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents selected from the group consisting of F, CI, Br, OH, =0 and OCH 3 ; preferably represents H or a d- 4 alkyl, which is unsubstituted; and

R 3 represents H, S(=0) 2 -NH 2 , a Ci -4 alkyl or a alkyl, wherein the Ci -4 alkyl is in each case unsubstituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents selected from the group consisting of F, CI, Br, OH, =0 and OCH 3 ;

preferably represents H, S(=0) 2 -NH 2 , a Ci -4 alkyl or a alkyl, wherein the Ci -4 alkyl is in each case unsubstituted; or - with the proviso that q is≠ 0 -

R 2 and R 3 together with the nitrogen atom connecting them form a 3 to 6 membered heterocyclyl, which is unsusbtituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents independently of one another selected from the group consisting of F, CI, Br, CN, CF 3 , CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , tert.-butyl, cyclopropyl, OH, =0, OCH 3 , OCF 3 , NH 2 , NH(CH 3 ) and N(CH 3 ) 2 ;

preferably together with the nitrogen atom connecting them form a 3 to 6 membered heterocyclyl, which is unsubstituted; or

A denotes CH or C(CH 3 ), and

R 2 represents H or a Ci -4 alkyl, which is unsubstituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents selected from the group consisting of F, CI, Br, OH, =0 and OCH 3 ; preferably represents H or a C 1-4 alkyl, which is unsubstituted; and

R 3 represents H, S(=0) 2 -NH 2 , a Ci -4 alkyl, which is unsubstituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents selected from the group consisting of F, CI, Br, OH, =0 and OCH 3 ;

preferably H, S(=0) 2 -NH 2 , or a Ci -4 alkyl, which is unsubstituted; or - with the proviso that q is≠ 0 -

R 2 and R 3 together with the nitrogen atom connecting them form a 3 to 6 membered heterocyclyl, which is unsusbtituted or mono-, di- or trisubstituted with 1 , 2 or 3 substituents independently of one another selected from the group consisting of F, CI, Br, CN, CF 3 , CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , tert.-butyl, cyclopropyl, OH, =0, OCH 3 , OCF 3 , NH 2 , NH(CH 3 ) and N(CH 3 ) 2 ;

preferably together with the nitrogen atom connecting them form a 3 to 6 membered heterocyclyl, which is unsubstituted.

In another preferred embodiment of the compound according to the present invention q denotes 0, 1 or 2, preferably 1 or 2, more preferably 1 , A denotes N,

R 0 is selected from the group consisting of H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ) and N(CH 3 ) 2 ; preferably is selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OCH 3 , OCF 3 , CH 3 , 0-CH 3 , 0-CH 2 CH 3 and N(CH 3 ) 2 ; more preferably is selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , OCF 3 , CH 3 , 0-CH 3 , and 0-CH 2 CH 3 ; even more preferably is selected from the group consisting of H, F, CI, CF 3 , OCF 3 , CH 3 and O- CH 3 ; still more preferably is selected from the group consisting of H, F, CI, CF 3 and 0-CH 3 ; in particular is selected from the group consisting of H, F, CI, and 0-CH 3 ; most preferred denotes F or CI; and

R 02 and R 03 are independently of one another selected from the group consisting of

H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ) and N(CH 3 ) 2 ; preferably are independently of one another selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , CN, CH 2 - OCH 3 , OCF 3 , CH 3 , 0-CH 3 , 0-CH 2 CH 3 and N(CH 3 ) 2 ; more preferably are independently of one another selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , OCF 3 , CH 3 , O- CH 3 , and 0-CH 2 CH 3 ; even more preferably are independently of one another selected from the group consisting of H, F, CI, CF 3 , OCF 3 , CH 3 and 0-CH 3 ; still more preferably are independently of one another selected from the group consisting of H, F, CI, CF 3 and 0-CH 3 ; in particular are independently of one another selected from the group consisting of H, F, CI, and 0-CH 3 ; most preferred denote independently of one another F or CI; or q denotes 1 or 2, preferably 1 ,

A denotes CH or C(CH 3 ), preferably C(CH 3 ),

R 0 is selected from the group consisting of H, F, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ) and N(CH 3 ) 2 ; preferably is selected from the group consisting of H, F, CFH 2 , CF 2 H, CF 3 , CN, CH 2 - OCH 3 , OCF 3 , CH 3 , 0-CH 3 , 0-CH 2 CH 3 and N(CH 3 ) 2 ; more preferably is selected from the group consisting of H, F, CFH 2 , CF 2 H, CF 3 , OCF 3 , CH 3 , 0-CH 3 , and 0-CH 2 CH 3 ; even more preferably is selected from the group consisting of H, F, CF 3 , OCF 3 , CH 3 and 0-CH 3 ; still more preferably is selected from the group consisting of H, F, CF 3 and 0-CH 3 ; in particular is selected from the group consisting of H, F and 0-CH 3 ; most preferred denotes F; and

R 02 and R 03 are independently of one another selected from the group consisting of

H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ) and N(CH 3 ) 2 ; preferably are independently of one another selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , CN, CH 2 - OCH 3 , OCF 3 , CH 3 , 0-CH 3 , 0-CH 2 CH 3 and N(CH 3 ) 2 ; more preferably are independently of one another selected from the group consisting of H, F, CI, CFH 2 , CF 2 H, CF 3 , OCF 3 , CH 3 , O- CH 3 , and 0-CH 2 CH 3 ; even more preferably are independently of one another selected from the group consisting of H, F, CI, CF 3 , OCF 3 , CH 3 and 0-CH 3 ; still more preferably are independently of one another selected from the group consisting of H, F, CI, CF 3 and 0-CH 3 ; in particular are independently of one another selected from the group consisting of H, F, CI, and 0-CH 3 ; most preferred denote independently of one another F or CI.

In a further preferred embodiment of the compound according to the present invention the part structure (RS1 )

q

(RS1 )

represents the part structure (PR1 )

(PR1 ),

wherein

R m represents NH 2 or an unsubstituted d -4 alkyl; preferably represents NH 2 , CH 3 or CH 2 CH 3 , more preferably represents NH 2 or CH 3 , in particular represents CH 3 , or represents the part structure (PR2-a) or (PR2-b) "C -4 alkyl

(PR2-a), (PR2-b), wherein the Ci_ 4 alkyl in part structure (PR2-b) is unsubstituted or monosubsituted with =0 or OH, preferably is unsubstituted, and wherein the Ci_ 4 alkyl in part structure (PR2-b) is preferably selected from the group consisting of methyl and ethyl, or represents one of the following part structures

Preferably, the part structure (RS1 )

q

(RS1 ) represents the part structure (PR1 )

(PR1 ),

wherein

R m represents NH 2 or an unsubstituted d -4 alkyl; preferably represents NH 2 , CH 3 or CH 2 CH 3 , more preferably represents NH 2 or CH 3 , in particular represents CH 3 .

In a particularly preferred embodiment of the present invention,

A denotes N and R 0 is selected from the group consisting of H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ) and N(CH 3 ) 2 , and

R 02 and R 03 are independently of one another selected from the group consisting of

H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ) and N(CH 3 ) 2 , preferably, wherein at least one of R 0 , R 02 and R 03 is≠ H, or

A denotes CH or C(CH 3 ), preferably C(CH 3 ), and

R 0 is selected from the group consisting of H, F, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ) and N(CH 3 ) 2 , and

R 02 and R 03 are independently of one another selected from the group consisting of

H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ) and N(CH 3 ) 2 ; preferably, wherein at least one of R 101 , R 02 and R 03 is≠ H,

R 2 represents CF 3 , tert.-butyl or cyclopropyl,

R 7 and R 9 are independently of one another selected from the group consisting of H, F, CI, Br, CF 3 , CN, OH, OCF 3 , CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , and 0-CH 2 CH 3 , preferably, wherein at least one of R 7 and R 9 is≠ H, the part structure (RS1 )

R 1 12

^ N. R1 13

q

(RS1 ) represents the part structure (PR1 )

(PR1 ),

wherein

R m represents NH 2 , CH 3 or CH 2 CH 3 .

In another particularly preferred embodiment of the present invention,

R 0 is selected from the group consisting of H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ) and N(CH 3 ) 2 , and

R 02 and R 03 are independently of one another selected from the group consisting of

H, F, CI, Br, CFH 2 , CF 2 H, CF 3 , CN, CH 2 -OH, CH 2 -OCH 3 , OCF 3 , OH, CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , 0-CH 2 CH 3 , NH 2 , NH(CH 3 ) and N(CH 3 ) 2 , preferably, wherein at least one of R 101 , R 02 and R 03 is≠ H,

R 2 represents CF 3 , tert.-butyl or cyclopropyl,

R 7 and R 9 are independently of one another selected from the group consisting of H, F, CI, Br, CF 3 , CN, OH, OCF 3 , CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , 0-CH 3 , and 0-CH 2 CH 3 , preferably, wherein at least one of R 7 and R 9 is≠ H,

A denotes N, and

R 2 represents H or a d -4 alkyl, which is unsubstituted, preferably denotes H, CH 3 or CH 2 CH 3 , more preferably denotes H or CH 3 , in particular denotes H,

R 3 represents H, S(=0) 2 -NH 2 , a Ci -4 alkyl or a alkyl, wherein the Ci -4 alkyl is in each case unsubstituted, preferably represents S(=0) 2 -NH 2 , or an unsubsitited alkyl, more preferably represents S(=0) 2 -NH 2 , S(=0) 2 -CH 3 or S(=0) 2 -CH 2 CH 3, even more preferably represents S(=0) 2 -NH 2 or S(=0) 2 -CH 3 , in particular represents S(=0) 2 -CH 3 , or

A denotes CH or C(CH 3 ), preferably C(CH 3 ),

R 2 represents H or a Ci -4 alkyl, which is unsubstituted, preferably denotes H, CH 3 or CH 2 CH 3 , more preferably denotes H or CH 3 , in particular denotes H,

R 3 represents H, S(=0) 2 -NH 2 , or a Ci -4 alkyl, which is unsubstituted, preferably represents S(=0) 2 -NH 2 .

Preferred embodiments of the compound according to the invention of general formula (R) have general formulae (RO-a) and/or (RO-b):

(RO-a) (RO-b), wherein the particular radicals, variables and indices have the meanings described herein in connection with the compounds according to the invention and preferred embodiments thereof.

Further preferred embodiments of the compound according to the invention of general formula (R) have general formulae (R1 -a), (R1 -a-1 ) and/or (R1 -a-2):

(R1 -a),

(FM -a-1 ),

(R1 -a-2), wherein the particular radicals, variables and indices have the meanings described herein in connection with the compounds according to the invention and preferred embodiments thereof.

Moreover, preferred embodiments of the compound according to the invention of general formula (R) have general formulae (R1 -b), (R1 -b-1 ) and/or (R1 -b-2):

(FM -b-1 ),

(FM -b-2), wherein the particular radicals, variables and indices have the meanings described herein in connection with the compounds according to the invention and preferred embodiments thereof.

In addition, preferred embodiments of the compound according to the invention of general formula (R) have general formulae (R1 -c), (R1 -c-1 ) and/or (R1 -c-2):

(R1 -C-1 ), (R1 -C-2), wherein the particular radicals, variables and indices have the meanings described herein in connection with the compounds according to the invention and preferred embodiments thereof. Yet further preferred embodiments of the compound according to the invention of general formula (R) have general formulae (R1 -d), (R1 -d-1 ) and/or (R1 -d-2):

(FM -d-1 ),

(FM -d-2), wherein the particular radicals, variables and indices have the meanings described herein in connection with the compounds according to the invention and preferred embodiments thereof.

Further preferred embodiments of the compound according to the invention of general formula (R) have general formulae (R1 -e), (R1 -e-1 ) and/or (R1 -e-2):

(RI -e-1 ), (R1 - wherein the particular radicals, variables and indices have the meanings described herein in connection with the compounds according to the invention and preferred embodiments thereof.

Additionally, preferred embodiments of the compound according to the invention of general formula (R) have general formulae (R1 -f), (R1 -f-1 ) and/or (R1 -f-2):

(FM -f-1 ), H

R 103 C 1-4 alkyl

R 102

(FM-f-2), wherein the particular radicals, variables and indices have the meanings described herein in connection with the compounds according to the invention and preferred embodiments thereof.

In particularly preferred embodiments of the present invention radical R 0 in the compound of general formula (R), (R1-a), (R1-a-1), (R1-b), (R1-b-1), (R1-c), (R1-C-1), (R1-d) and/or (R1-d-1) represents F, CI, CF 3 or 0-CH 3 , preferably F or CI, most preferably CI - preferably when R 03 is H and R 02 represents H, F, CI, CF 3 or OCH 3 , more preferably when R 03 is H and R 02 represents H, F or CI, even more preferably when both R 02 and R 03 denote H, and the remaining particular radicals, variables and indices have the meanings described herein in connection with the compounds according to the invention and preferred embodiments thereof.

In particularly preferred embodiments of the present invention radical R 0 in the compound of general formula (R), (R1-a), (R1-a-2), (R1-b), (R1-b-2), (R1-c), (R1-C-2), (R1-d) and/or (R1-d-2), represents F, CF 3 or 0-CH 3 , preferably F or OCH 3 , most preferably F - preferably when R 03 is H and R 02 represents H, F, CI, CF 3 or OCH 3 , more preferably when R 03 is H and R 02 represents H, F or CI, even more preferably when both R 02 and R 03 denote H, and the remaining particular radicals, variables and indices have the meanings described herein in connection with the compounds according to the invention and preferred embodiments thereof.

In further particularly preferred embodiments of the present invention radical R 0 in the compound of general formula (R1-e), (R1-e-1), (R1-e-2), (R1-f), (R1-f-1) and/or (R1-f-2) represents F, CI, CF 3 or 0-CH 3 , preferably F or CI, most preferably CI - preferably when R 03 is H and R 02 represents H, F, CI, CF 3 or OCH 3 , more preferably when R 03 is H and R 02 represents H, F or CI, even more preferably when both R 02 and R 03 denote H, and the remaining particular radicals, variables and indices have the meanings described herein in connection with the compounds according to the invention and preferred embodiments thereof.

Particularly preferred are compounds according to the invention from the group

B1 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B2 N-[[2-(3-Chloro-4-fluoro-phenyl)-5-(trifluoromethyl)-2H-pyra zol-3-yl]-methyl]-2-[3- fluoro-4-(methanesulfonamido-methyl)-phenyl]-propionamide;

B3 N-[[5-tert-Butyl-2-(3-chloro-4-fluoro-phenyl)-2H-pyrazol-3-y l]-methyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B4 N-[[2-(3-Chlorophenyl)-5-cyclopropyl-2H-pyrazol-3-yl]-methyl ]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B5 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B6 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B7 2-[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-N-[[2-(3-f luorophenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-propionamide;

B8 2-[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-N-[[5-(tri fluoromethyl)-2-[3-

(trifluoromethyl)phenyl]-2H-pyrazol-3-yl]-methyl]-propion amide;

B9 N-[[5-tert-Butyl-2-(3-fluorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B10 N-[[5-tert-Butyl-2-(3,4-difluoro-phenyl)-2H-pyrazol-3-yl]-me thyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B11 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[4-

(methanesulfonamido-methyl)-3-methoxy-phenyl]-urea;

B12 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[4-

(methanesulfonamido-methyl)-3-methoxy-phenyl]-urea;

B13 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[3-chloro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B14 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[4-(methylamino- methyl)-phenyl]-urea;

B15 2-[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-N-[[2-(3-m ethoxyphenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-propionamide;

B16 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[4-

[[(ethylsulfonyl)amino]-methyl]-3-fluoro-phenyl]-propiona mide; B17 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[4-

[[(ethylsulfonyl)amino]-methyl]-3-fluoro-phenyl]-propiona mide;

B18 N-[[2-(4-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B19 N-[[2-(3,4-Difluoro-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3 -yl]-methyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B20 N-[[5-tert-Butyl-2-(4-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B21 N-[[5-tert-Butyl-2-(4-fluorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B22 2-[3-Chloro-4-(methanesulfonamido-methyl)-phenyl]-N-[[2-(3-c hlorophenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-propionamide;

B23 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-acetamide;

B24 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[4-(ethylamino- methyl)-3-fluoro-phenyl]-urea;

B25 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[4-(ethylamino-methyl)-

3-fluoro-phenyl]-urea;

B26 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-acetamide;

B27 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[3,5-difluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B28 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[4-

(methanesulfonamido-methyl)-phenyl]-urea;

B29 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[3-fluoro-4-

[(methyl-methylsulfonyl-amino)-methyl]-phenyl]-propionami de;

B30 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[3-fluoro-4-[(methyl- methylsulfonyl-amino)-methyl]-phenyl]-propionamide;

B31 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B32 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[3,5-difluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B33 N-[[4-[[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3 -yl]-methyl- carbamoyl]amino]-2-fluoro-phenyl]-methyl]-acetamide;

B34 N-[[4-[[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-me thyl-carbamoyl]amino]-2- fluoro-phenyl]-methyl]-acetamide; B35 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B36 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[4-[(sulfamoylamino)- methyl]-phenyl]-urea;

B37 2-[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-N-[[2-(3-f luorophenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-acetamide;

B38 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[4-

(dimethylaminomethyl)-3-fluoro-phenyl]-urea;

B39 1 -[4-(Aminomethyl)-3-fluoro-phenyl^

pyrazol-3-yl]-methyl]-urea;

B40 1 -[4-(Aminomethyl)-3-fluoro-phenyl]-3-[[5-tert-butyl-2-(3-chl orophenyl)-2H-pyrazol-3- yl]-methyl]-urea;

B41 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B42 2-[4-(Aminomethyl)-3-fluoro-phenyl]-N-[[2-(3-chlorophenyl)-5 -(trifluorome

pyrazol-3-yl]-methyl]-propionamide;

B43 N-[[5-tert-Butyl-2-[3-(trifluoromethyl)phenyl]-2H-pyrazol-3- yl]-methyl]-2-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B44 1 -[[2-(3-Fluorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[4-

(methanesulfonamido-methyl)-phenyl]-urea;

B45 1 -[[5-tert-Butyl-2-(3-fluorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[4-(methanesulfonamido- methyl)-phenyl]-urea;

B46 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B47 2-[4-(Aminomethyl)-3-fluoro-phenyl]-N-[[5-tert-butyl-2-(3-ch lorophenyl)-2H-pyrazol-3- yl]-methyl]-propionamide;

B48 1 -[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-3-[[2-(3-fl uorophenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea;

B49 1 -[[2-(3,4-Difluoro-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3- yl]-methyl]-3-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B50 1 -[[5-tert-Butyl-2-(3-fluorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B51 1 -[[5-tert-Butyl-2-(3-chloro-4-fluoro-phenyl)-2H-pyrazol-3-yl ]-methyl]-3-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B52 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea; B53 1 -[[5-tert-Butyl-2-(4-fluorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B54 1 -[[5-tert-Butyl-2-[3-(trifluoromethyl)phenyl]-2H-pyrazol-3-y l]-methyl]-3-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B55 1 -[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-3-[[5-(trif luoromethyl)-2-[3-

(trifluoromethyl)phenyl]-2H-pyrazol-3-yl]-methyl]-urea;

B56 1 -[[5-tert-Butyl-2-(3-fluorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[4-[(sulfamoylamino)- methyl]-phenyl]-urea;

B57 1 -[[2-(3,4-Difluoro-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3- yl]-methyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B58 1 -[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-3-[[2-(3-me thoxyphenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea;

B59 1 -[[5-tert-Butyl-2-(3-fluorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B60 1 -[[2-(3-Fluorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B61 1 -[[2-(3-Chlorophenyl)-5-cyclopropyl-2H-pyrazol-3-yl]-methyl] -3-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B62 1 -[[2-(3,4-Difluoro-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3- yl]-methyl]-3-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B63 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[3-fluoro-4-

(methylamino-methyl)-phenyl]-propionamide;

B64 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[4-

(dimethylaminomethyl)-3-fluoro-phenyl]-propionamide;

B65 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[4-

(dimethylaminomethyl)-3-fluoro-phenyl]-propionamide;

B66 2-[4-(Acetylamino-methyl)-3-fluoro-phenyl]-N-[[2-(3-chloroph enyl)-5-(trifluoromethyl)-

2H-pyrazol-3-yl]-methyl]-propionamide;

B67 2-[4-(Acetylamino-methyl)-3-fluoro-phenyl]-N-[[5-tert-butyl- 2-(3-chlorophenyl)-2H- pyrazol-3-yl]-methyl]-propionamide;

B68 2-[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-N-[[2-(m-t olyl)-5-(trifluoromethyl)-

2H-pyrazol-3-yl]-methyl]-propionamide;

B69 1 -[[5-tert-Butyl-2-(3,4-difluoro-phenyl)-2H-pyrazol-3-yl]-met hyl]-3-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B70 1 -[[2-(3-Chloro-4-fluoro-phenyl)-5-(trifluoromethyl)-2H-pyraz ol-3-yl]-methyl]-3-[3- fluoro-4-[(sulfamoylamino)-methyl]-phenyl]-urea; B71 1 -[3-Fluoro-4-[(sulfamoylamino)-methyl]-phenyl]-3-[[2-(3-meth oxyphenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea;

B72 1 -[[5-tert-Butyl-2-(3,4-difluoro-phenyl)-2H-pyrazol-3-yl]-met hyl]-3-[3-fluoro-4-

(methanesulfonamido-methyl)-phenyl]-urea;

B73 1 -[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-3-[[2-(3-is opropyl-phenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea;

B74 1 -[[5-tert-Butyl-2-(3,4-difluoro-phenyl)-2H-pyrazol-3-yl]-met hyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B75 1 -[[2-(3-Chlorophenyl)-5-cyclopropyl-2H-pyrazol-3-yl]-methyl] -3-[4-[(sulfamoylamino)- methyl]-phenyl]-urea;

B76 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[4-

(methanesulfonamido-methyl)-3-methoxy-phenyl]-propionamid e;

B77 1 -[[2-(3-Fluorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B78 1 -[[2-(3-Chloro-4-fluoro-phenyl)-5-(trifluoromethyl)-2H-pyraz ol-3-yl]-methyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B79 1 -[[2-(3-lsopropyl-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3-y l]-methyl]-3-[4-

[(sulfamoylamino)-methyl]-phenyl]-urea;

B80 1 -[3-Fluoro-4-[(sulfamoylamino)-methyl]-phenyl]-3-[[2-(3-isop ropyl-phenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea;

B81 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[4-

(methanesulfonamido-methyl)-phenyl]-urea;

B82 2-[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-N-[[2-(3-i sopropyl-phenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-propionamide;

B83 1 -[[2-(3-lsopropyl-phenyl)-5-(trifluoromethyl)-2H-pyrazol-3-y l]-methyl]-3-[4-

(methanesulfonamido-methyl)-phenyl]-urea;

B84 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[4-

[[(ethylsulfonyl)amino]-methyl]-phenyl]-urea;

B85 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[4-

[[(ethylsulfonyl)amino]-methyl]-phenyl]-urea;

B86 1 -[4-(Methanesulfonamido-methyl)-3-methoxy-phenyl]-3-[[2-(m-t olyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea;

B87 1 -[3-Fluoro-4-(methanesulfonamido-methyl)-phenyl]-3-[[2-(m-to lyl)-5-(trifluoromethyl)-

2H-pyrazol-3-yl]-methyl]-urea;

B88 N-[[5-tert-Butyl-2-(m-tolyl)-2H-pyrazol-3-yl]-methyl]-2-[3-f luoro-4-

(methanesulfonamido-methyl)-phenyl]-propionamide; B89 1 -[4-(Methanesulfonamido-methyl)-phenyl]-3-[[2-(m-tolyl)-5-(t rifluoromethyl)-2H- pyrazol-3-yl]-methyl]-urea;

B90 1 -[[5-tert-Butyl-2-(m-tolyl)-2H-pyrazol-3-yl]-methyl]-3-[4-(m ethanesulfonamido- methyl)-phenyl]-urea;

B91 1 -[4-[[(Ethylsulfonyl)amino]-methyl]-phenyl]-3-[[2-(m-tolyl)- 5-(trifluoromethyl)-2H- pyrazol-3-yl]-methyl]-urea;

B92 1 -[[5-tert-Butyl-2-(m-tolyl)-2H-pyrazol-3-yl]-methyl]-3-[4-[[ (ethylsulfonyl)amino]- methyl]-phenyl]-urea;

B93 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-propionamide;

B94 2-[3-Fluoro-4-[(sulfamoylamino)-methyl]-phenyl]-N-[[2-(3-iso propyl-phenyl)-5-

(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-propionamide;

B95 N-[[2-(3-Chlorophenyl)-5-cyclopropyl-2H-pyrazol-3-yl]-methyl ]-2-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-propionamide;

B96 N-[[5-tert-Butyl-2-(3-chloro-4-fluoro-phenyl)-2H-pyrazol-3-y l]-methyl]-2-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-propionamide;

B97 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[3-fluoro-4-

[(sulfamoylamino)-methyl]-phenyl]-propionamide;

B98 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[4-

(methanesulfonamido-methyl)-3-methoxy-phenyl]-propionamid e;

B99 1 -[[2-(3-Chlorophenyl)-5-(trifluorom

methyl)-phenyl]-urea;

B100 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[3-fluoro-4-

(piperidin-1 -yl-methyl)-phenyl]-urea;

B101 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[3-fluoro-4-(pyrrolidin-1 - yl-methyl)-phenyl]-urea;

B102 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[4-

[(sulfamoylamino)-methyl]-phenyl]-propionamide;

B103 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[4-[(sulfamoylamino)- methyl]-phenyl]-propionamide;

B104 N-[[2-(3-Fluorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[4-

(methanesulfonamido-methyl)-phenyl]-propionamide;

B105 1 -[[2-(3-Fluorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-m ethyl]-3-[4-

(methanesulfonamido-methyl)-3-methoxy-phenyl]-urea;

B106 N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[4-

(methanesulfonamido-methyl)-3-methyl-phenyl]-propionamide ; and B107 N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[4- (methanesulfonamido-methyl)-3-methyl-phenyl]-propionamide; optionally in the form of a single stereoisomer or a mixture of stereoisomers, in the form of the free compound and/or a physiologically acceptable salt thereof.

Furthermore, preference may be given to compounds according to the invention that cause a 50% displacement of capsaicin, which is present at a concentration of 100 nM, in a FLIPR assay with CHO K1 cells which were transfected with the human VR1 gene at a concentration of less than 2 000 nM, preferably less than 1 000 nM, particularly preferably less than 300 nM, most particularly preferably less than 100 nM, even more preferably less than 75 nM, additionally preferably less than 50 nM, most preferably less than 10 nM.

In the process, the Ca 2+ influx is quantified in the FLIPR assay with the aid of a Ca 2+ - sensitive dye (type Fluo-4, Molecular Probes Europe BV, Leiden, the Netherlands) in a fluorescent imaging plate reader (FLIPR, Molecular Devices, Sunnyvale, USA), as described hereinafter.

The substituted compounds according to the invention and corresponding stereoisomers and also the respective corresponding acids, bases, salts and solvates are toxicologically safe and are therefore suitable as pharmaceutical active ingredients in pharmaceutical compositions.

The present invention therefore further relates to a pharmaceutical composition containing at least one compound according to the invention, in each case if appropriate in the form of one of its pure stereoisomers, in particular enantiomers or diastereomers, its racemates or in the form of a mixture of stereoisomers, in particular the enantiomers and/or diastereomers, in any desired mixing ratio, or respectively in the form of a corresponding salt, or respectively in the form of a corresponding solvate, and also if appropriate one or more pharmaceutically compatible auxiliaries.

These pharmaceutical compositions according to the invention are suitable in particular for vanilloid receptor 1 -(VR1/TRPV1 ) regulation, preferably for vanilloid receptor 1 -(VR1/TRPV1 ) inhibition and/or for vanilloid receptor 1 -(VR1 /TRPV1 ) stimulation, i.e. they exert an agonistic or antagonistic effect. Likewise, the pharmaceutical compositions according to the invention are preferably suitable for the prophylaxis and/or treatment of disorders or diseases which are mediated, at least in part, by vanilloid receptors 1 .

The pharmaceutical composition according to the invention is suitable for administration to adults and children, including toddlers and babies.

The pharmaceutical composition according to the invention may be found as a liquid, semisolid or solid pharmaceutical form, for example in the form of injection solutions, drops, juices, syrups, sprays, suspensions, tablets, patches, capsules, plasters, suppositories, ointments, creams, lotions, gels, emulsions, aerosols or in multiparticulate form, for example in the form of pellets or granules, if appropriate pressed into tablets, decanted in capsules or suspended in a liquid, and also be administered as much.

In addition to at least one substituted compound according to the invention, if appropriate in the form of one of its pure stereoisomers, in particular enantiomers or diastereomers, its racemate or in the form of mixtures of the stereoisomers, in particular the enantiomers or diastereomers, in any desired mixing ratio, or if appropriate in the form of a corresponding salt or respectively in the form of a corresponding solvate, the pharmaceutical composition according to the invention conventionally contains further physiologically compatible pharmaceutical auxiliaries which can for example be selected from the group consisting of excipients, fillers, solvents, diluents, surface-active substances, dyes, preservatives, blasting agents, slip additives, lubricants, aromas and binders.

The selection of the physiologically compatible auxiliaries and also the amounts thereof to be used depend on whether the pharmaceutical composition is to be applied orally, subcutaneously, parenterally, intravenously, intraperitoneal^, intradermal^, intramuscularly, intranasally, buccally, rectally or locally, for example to infections of the skin, the mucous membranes and of the eyes. Preparations in the form of tablets, dragees, capsules, granules, pellets, drops, juices and syrups are preferably suitable for oral application; solutions, suspensions, easily reconstitutable dry preparations and also sprays are preferably suitable for parenteral, topical and inhalative application. The substituted compounds according to the invention used in the pharmaceutical composition according to the invention in a repository in dissolved form or in a plaster, agents promoting skin penetration being added if appropriate, are suitable percutaneous application preparations. Orally or percutaneously applicable preparation forms can release the respective substituted compound according to the invention also in a delayed manner. The pharmaceutical compositions according to the invention are prepared with the aid of conventional means, devices, methods and process known in the art, such as are described for example in„Remington's Pharmaceutical Sciences", A.R. Gennaro (Editor), 17 th edition, Mack Publishing Company, Easton, Pa, 1985, in particular in Part 8, Chapters 76 to 93. The corresponding description is introduced herewith by way of reference and forms part of the disclosure. The amount to be administered to the patient of the respective substituted compounds according to the invention of the above-indicated general formula I may vary and is for example dependent on the patient's weight or age and also on the type of application, the indication and the severity of the disorder. Conventionally 0.001 to 100 mg/kg, preferably 0.05 to 75 mg/kg, particularly preferably 0.05 to 50 mg of at least one such compound according to the invention are applied per kg of the patient's body weight.

The pharmaceutical composition according to the invention is preferably suitable for the treatment and/or prophylaxis of one or more disorders and/or diseases selected from the group consisting of pain, preferably pain selected from the group consisting of acute pain, chronic pain, neuropathic pain, visceral pain and joint pain; hyperalgesia; allodynia; causalgia; migraine; depression; nervous affection; axonal injuries; neurodegenerative diseases, preferably selected from the group consisting of multiple sclerosis, Alzheimer's disease, Parkinson's disease and Huntington's disease; cognitive dysfunctions, preferably cognitive deficiency states, particularly preferably memory disorders; epilepsy; respiratory diseases, preferably selected from the group consisting of asthma, bronchitis and pulmonary inflammation; coughs; urinary incontinence; overactive bladder (OAB); disorders and/or injuries of the gastrointestinal tract; duodenal ulcers; gastric ulcers; irritable bowel syndrome; strokes; eye irritations; skin irritations; neurotic skin diseases; allergic skin diseases; psoriasis; vitiligo; herpes simplex; inflammations, preferably inflammations of the intestine, the eyes, the bladder, the skin or the nasal mucous membrane; diarrhoea; pruritus; osteoporosis; arthritis; osteoarthritis; rheumatic diseases; eating disorders, preferably selected from the group consisting of bulimia, cachexia, anorexia and obesity; medication dependency; misuse of medication; withdrawal symptoms in medication dependency; development of tolerance to medication, preferably to natural or synthetic opioids; drug dependency; misuse of drugs; withdrawal symptoms in drug dependency; alcohol dependency; misuse of alcohol and withdrawal symptoms in alcohol dependency; for diuresis; for antinatriuresis; for influencing the cardiovascular system; for increasing vigilance; for the treatment of wounds and/or burns; for the treatment of severed nerves; for increasing libido; for modulating movement activity; for anxiolysis; for local anaesthesia and/or for inhibiting undesirable side effects, preferably selected from the group consisting of hyperthermia, hypertension and bronchoconstriction, triggered by the administration of vanilloid receptor 1 (VR1 /TRPV1 receptor) agonists, preferably selected from the group consisting of capsaicin, resiniferatoxin, olvanil, arvanil, SDZ-249665, SDZ-249482, nuvanil and capsavanil.

Particularly preferably, the pharmaceutical composition according to the invention is suitable for the treatment and/or prophylaxis of one or more disorders and/or diseases selected from the group consisting of pain, preferably of pain selected from the group consisting of acute pain, chronic pain, neuropathic pain, visceral pain and joint pain; migraine; depression; neurodegenerative diseases, preferably selected from the group consisting of multiple sclerosis, Alzheimer's disease, Parkinson's disease and Huntington's disease; cognitive dysfunctions, preferably cognitive deficiency states, particularly preferably memory disorders; inflammations, preferably inflammations of the intestine, the eyes, the bladder, the skin or the nasal mucous membrane; urinary incontinence; overactive bladder (OAB); medication dependency; misuse of medication; withdrawal symptoms in medication dependency; development of tolerance to medication, preferably development of tolerance to natural or synthetic opioids; drug dependency; misuse of drugs; withdrawal symptoms in drug dependency; alcohol dependency; misuse of alcohol and withdrawal symptoms in alcohol dependency.

Most particularly preferably, the pharmaceutical composition according to the invention is suitable for the treatment and/or prophylaxis of pain, preferably of pain selected from the group consisting of acute pain, chronic pain, neuropathic pain and visceral pain.

The present invention further relates to a substituted compound according to the present invention and also if appropriate to a substituted compound according to the present invention and one or more pharmaceutically acceptable auxiliaries for use in vanilloid receptor 1 -(VR1 /TRPV1 ) regulation, preferably for use in vanilloid receptor 1 -(VR1/TRPV1 ) inhibition and/or vanilloid receptor 1 -(VR1 /TRPV1 ) stimulation.

The present invention therefore further relates to a substituted compound according to the present invention and also if appropriate to a substituted compound according to the present invention and one or more pharmaceutically acceptable auxiliaries for use in the prophylaxis and/or treatment of disorders and/or diseases which are mediated, at least in part, by vanilloid receptors 1 . In particular, the present invention therefore further relates to a substituted compound according to the present invention and also if appropriate to a substituted compound according to the present invention and one or more pharmaceutically acceptable auxiliaries for use in the prophylaxis and/or treatment of disorders and/or diseases selected from the group consisting of pain, preferably pain selected from the group consisting of acute pain, chronic pain, neuropathic pain, visceral pain and joint pain; hyperalgesia; allodynia; causalgia; migraine; depression; nervous affection; axonal injuries; neurodegenerative diseases, preferably selected from the group consisting of multiple sclerosis, Alzheimer's disease, Parkinson's disease and Huntington's disease; cognitive dysfunctions, preferably cognitive deficiency states, particularly preferably memory disorders; epilepsy; respiratory diseases, preferably selected from the group consisting of asthma, bronchitis and pulmonary inflammation; coughs; urinary incontinence; overactive bladder (OAB); disorders and/or injuries of the gastrointestinal tract; duodenal ulcers; gastric ulcers; irritable bowel syndrome; strokes; eye irritations; skin irritations; neurotic skin diseases; allergic skin diseases; psoriasis; vitiligo; herpes simplex; inflammations, preferably inflammations of the intestine, the eyes, the bladder, the skin or the nasal mucous membrane; diarrhoea; pruritus; osteoporosis; arthritis; osteoarthritis; rheumatic diseases; eating disorders, preferably selected from the group consisting of bulimia, cachexia, anorexia and obesity; medication dependency; misuse of medication; withdrawal symptoms in medication dependency; development of tolerance to medication, preferably to natural or synthetic opioids; drug dependency; misuse of drugs; withdrawal symptoms in drug dependency; alcohol dependency; misuse of alcohol and withdrawal symptoms in alcohol dependency; for diuresis; for antinatriuresis; for influencing the cardiovascular system; for increasing vigilance; for the treatment of wounds and/or burns; for the treatment of severed nerves; for increasing libido; for modulating movement activity; for anxiolysis; for local anaesthesia and/or for inhibiting undesirable side effects, preferably selected from the group consisting of hyperthermia, hypertension and bronchoconstriction, triggered by the administration of vanilloid receptor 1 (VR1 /TRPV1 receptor) agonists, preferably selected from the group consisting of capsaicin, resiniferatoxin, olvanil, arvanil, SDZ-249665, SDZ-249482, nuvanil and capsavanil.

Most particularly preferred is a substituted compound according to the present invention and also if appropriate to a substituted compound according to the present invention and one or more pharmaceutically acceptable auxiliaries for use in the prophylaxis and/or treatment of pain, preferably of pain selected from the group consisting of acute pain, chronic pain, neuropathic pain and visceral pain. The present invention further relates to the use of at least one substituted compound according to the present invention and also if appropriate to the use of at least one substituted compound according to the present invention of one or more pharmaceutically acceptable auxiliaries for the preparation of a pharmaceutical composition for vanilloid receptor 1 -(VR1 /TRPV1 ) regulation, preferably for vanilloid receptor 1 -(VR1/TRPV1 ) inhibition and/or for vanilloid receptor 1 -(VR1 /TRPV1 ) stimulation, and, further for the prophylaxis and/or treatment of disorders and/or diseases which are mediated, at least in part, by vanilloid receptors 1 , such as e.g. disorders and/or diseases selected from the group consisting of pain, preferably pain selected from the group consisting of acute pain, chronic pain, neuropathic pain, visceral pain and joint pain; hyperalgesia; allodynia; causalgia; migraine; depression; nervous affection; axonal injuries; neurodegenerative diseases, preferably selected from the group consisting of multiple sclerosis, Alzheimer's disease, Parkinson's disease and Huntington's disease; cognitive dysfunctions, preferably cognitive deficiency states, particularly preferably memory disorders; epilepsy; respiratory diseases, preferably selected from the group consisting of asthma, bronchitis and pulmonary inflammation; coughs; urinary incontinence; overactive bladder (OAB); disorders and/or injuries of the gastrointestinal tract; duodenal ulcers; gastric ulcers; irritable bowel syndrome; strokes; eye irritations; skin irritations; neurotic skin diseases; allergic skin diseases; psoriasis; vitiligo; herpes simplex; inflammations, preferably inflammations of the intestine, the eyes, the bladder, the skin or the nasal mucous membrane; diarrhoea; pruritus; osteoporosis; arthritis; osteoarthritis; rheumatic diseases; eating disorders, preferably selected from the group consisting of bulimia, cachexia, anorexia and obesity; medication dependency; misuse of medication; withdrawal symptoms in medication dependency; development of tolerance to medication, preferably to natural or synthetic opioids; drug dependency; misuse of drugs; withdrawal symptoms in drug dependency; alcohol dependency; misuse of alcohol and withdrawal symptoms in alcohol dependency; for diuresis; for antinatriuresis; for influencing the cardiovascular system; for increasing vigilance; for the treatment of wounds and/or burns; for the treatment of severed nerves; for increasing libido; for modulating movement activity; for anxiolysis; for local anaesthesia and/or for inhibiting undesirable side effects, preferably selected from the group consisting of hyperthermia, hypertension and bronchoconstriction, triggered by the administration of vanilloid receptor 1 (VR1 /TRPV1 receptor) agonists, preferably selected from the group consisting of capsaicin, resiniferatoxin, olvanil, arvanil, SDZ-249665, SDZ-249482, nuvanil and capsavanil.

Another aspect of the present invention is a method for vanilloid receptor 1 -(VR1/TRPV1 ) regulation, preferably for vanilloid receptor 1 -(VR1 /TRPV1 ) inhibition and/or for vanilloid receptor 1 -(VR1 /TRPV1 ) stimulation, and, further, a method of treatment and/or prophylaxis of disorders and/or diseases, which are mediated, at least in part, by vanilloid receptors 1 , in a mammal, preferably of disorders and/or diseases selected from the group consisting of pain, preferably pain selected from the group consisting of acute pain, chronic pain, neuropathic pain, visceral pain and joint pain; hyperalgesia; allodynia; causalgia; migraine; depression; nervous affection; axonal injuries; neurodegenerative diseases, preferably selected from the group consisting of multiple sclerosis, Alzheimer's disease, Parkinson's disease and Huntington's disease; cognitive dysfunctions, preferably cognitive deficiency states, particularly preferably memory disorders; epilepsy; respiratory diseases, preferably selected from the group consisting of asthma, bronchitis and pulmonary inflammation; coughs; urinary incontinence; overactive bladder (OAB); disorders and/or injuries of the gastrointestinal tract; duodenal ulcers; gastric ulcers; irritable bowel syndrome; strokes; eye irritations; skin irritations; neurotic skin diseases; allergic skin diseases; psoriasis; vitiligo; herpes simplex; inflammations, preferably inflammations of the intestine, the eyes, the bladder, the skin or the nasal mucous membrane; diarrhoea; pruritus; osteoporosis; arthritis; osteoarthritis; rheumatic diseases; eating disorders, preferably selected from the group consisting of bulimia, cachexia, anorexia and obesity; medication dependency; misuse of medication; withdrawal symptoms in medication dependency; development of tolerance to medication, preferably to natural or synthetic opioids; drug dependency; misuse of drugs; withdrawal symptoms in drug dependency; alcohol dependency; misuse of alcohol and withdrawal symptoms in alcohol dependency; for diuresis; for antinatriuresis; for influencing the cardiovascular system; for increasing vigilance; for the treatment of wounds and/or burns; for the treatment of severed nerves; for increasing libido; for modulating movement activity; for anxiolysis; for local anaesthesia and/or for inhibiting undesirable side effects, preferably selected from the group consisting of hyperthermia, hypertension and bronchoconstriction, triggered by the administration of vanilloid receptor 1 (VR1/TRPV1 receptor) agonists, preferably selected from the group consisting of capsaicin, resiniferatoxin, olvanil, arvanil, SDZ-249665, SDZ-249482, nuvanil and capsavanil, which comprises administering an effective amount of at least one substituted compound according to the invention to the mammal.

The effectiveness against pain can be shown, for example, in the Bennett or Chung model (Bennett, G.J. and Xie, Y.K., A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man, Pain 1988, 33(1 ), 87-107; Kim, S.H. and Chung, J.M., An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat, Pain 1992, 50(3), 355-363), by tail flick experiments (e.g. according to D'Amour und Smith (J. Pharm. Exp. Ther. 72, 74 79 (1941 )) or by the formalin test (e.g. according to D. Dubuisson et al., Pain 1977, 4, 161 -174).

The present invention further relates to processes for preparing substituted compounds according to the invention.

In particular, the compounds according to the present invention of can be prepared by a process according to which at least one compound of general formula (R-ll),

(R-ll), in which R 0 , R 02 , R 03 and R 2 have one of the foregoing meanings, is reacted in a reaction medium, if appropriate in the presence of at least one suitable coupling reagent, if appropriate in the presence of at least one base, with a compound of general formula (R-lll) with D = OH or Hal,

(R-lll), in which Hal represents a halogen, preferably Br or CI, and R 7 , R 9 , R 2 , R 3 and q each have one of the foregoing meanings and A denotes CH or C(CH 3 ), in a reaction medium, if appropriate in the presence of at least one suitable coupling reagent, if appropriate in the presence of at least one base, to form a compound of general formula (R),

(R). in which A represents CH or C(CH 3 ) and R 0 , R 02 , R 03 and R 2 as well as R 7 , R 9 , R 2 , R 3 and q have one of the foregoing meanings; or in that at least one compound of general formula (R- II),

R 1

(R-ll), in which R 0 , R 02 , R 03 and R 2 have one of the foregoing meanings, is reacted to form a compound of general formula (R-IV),

(R-IV), in which R 0 , R 02 , R 03 and R 2 have one of the foregoing meanings, in a reaction medium, in the presence of phenyl chloroformiate, if appropriate in the presence of at least one base and/or at least one coupling reagent, and said compound is if appropriate purified and/or isolated, and a compound of general formula (R-IV) is reacted with a compound of general formula (R-V),

(R-V), in which R 7 , R 9 , R 2 , R 3 and q have one of the foregoing meanings, and A denotes N, in a reaction medium, if appropriate in the presence of at least one suitable coupling reagent, if appropriate in the presence of at least one base, to form a compound of general formula (R),

(R). in which A represents N and R 0 , R 02 , R 03 and R 2 as well as R 7 , R 9 , R 2 , R 3 and q have one of the foregoing meanings.

The reaction of compounds of the above- indicated general formula (R-ll) with carboxylic acids of the above-indicated general formula (R-lll), particularly with D = OH, to form compounds of the above- indicated general formula (R) is carried out preferably in a reaction medium selected from the group consisting of diethyl ether, tetrahydrofuran, acetonitrile, methanol, ethanol, (1 ,2)-dichloroethane, dimethylformamide, dichloromethane and corresponding mixtures, if appropriate in the presence of at least one coupling reagent, preferably selected from the group consisting of l -benzotriazolyloxy-tris-(dimethylamino)- phosphonium hexafluorophosphate (BOP), dicyclohexylcarbodiimide (DCC), N'-(3- dimethylaminopropyl)-N-ethylcarbodiimide (EDCI), diisopropylcarbodiimide, 1 ,1 '- carbonyldiimidazole (CDI), N-[(dimethylamino)-1 H-1 , 2, 3-triazolo[4, 5-b]pyridino-1 -yl- methylene]-N-methylmethanaminium hexafluorophosphate N-oxide (HATU), 0-(benzotriazol- 1 -yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HBTU), O-(benzotriazol-l -yl)- Ν,Ν,Ν',Ν'-tetramethyluronium tetrafluoroborate (TBTU), N-hydroxybenzotriazole (HOBt) and 1 -hydroxy-7-azabenzotriazole (HOAt), if appropriate in the presence of at least one organic base, preferably selected from the group consisting of triethylamine, pyridine, dimethylaminopyridine, N-methylmorpholine and diisopropylethylamine, preferably at temperatures of from -70 °C to 100 °C.

Alternatively, the reaction of compounds of the above- indicated general formulae (R-ll) with carboxylic acid halides of the above-indicated general formula (R-ll I) with D = Hal, in which Hal represents a halogen as the leaving group, preferably a chlorine or bromine atom, to form compounds of the above-indicated general formula (R) is carried out in a reaction medium preferably selected from the group consisting of diethyl ether, tetrahydrofuran, acetonitrile, methanol, ethanol, dimethylformamide, dichloromethane and corresponding mixtures, if appropriate in the presence of an organic or inorganic base, preferably selected from the group consisting of triethylamine, dimethylaminopyridine, pyridine and diisopropylamine, at temperatures of from -70 ^ to 100 °C.

The compounds of the above- indicated formulae (R-ll), (R-lll), (R-IV), and (R-V) are each commercially available and/or can be prepared using conventional processes known to the person skilled in the art. In particular, processes to prepare these compounds are e.g. disclosed in WO 2010/127855-A2, and WO 2010/127856-A2. The corresponding parts of these references are hereby deemed to be part of the disclosure.

All reactions which can be applied for synthesizing the compounds according to the present invention can each be carried out under the conventional conditions with which the person skilled in the art is familiar, for example with regard to pressure or the order in which the components are added. If appropriate, the person skilled in the art can determine the optimum procedure under the respective conditions by carrying out simple preliminary tests. The intermediate and end products obtained using the reactions described hereinbefore can each be purified and/or isolated, if desired and/or required, using conventional methods known to the person skilled in the art. Suitable purifying processes are for example extraction processes and chromatographic processes such as column chromatography or preparative chromatography. All of the process steps of the reaction sequences which can be applied for synthesizing the compounds according to the present invention as well as the respective purification and/or isolation of intermediate or end products, can be carried out partly or completely under an inert gas atmosphere, preferably under a nitrogen atmosphere. The substituted compounds according to the invention can be isolated both in the form of their free bases, and also in the form of corresponding salts, in particular physiologically acceptable salts, and further in the form of a solvate such as hydrate.

The free bases of the respective substituted compounds according to the invention can be converted into the corresponding salts, preferably physiologically acceptable salts, for example by reaction with an inorganic or organic acid, preferably with hydrochloric acid, hydrobromic acid, sulphuric acid, methanesulphonic acid, p-toluenesulphonic acid, carbonic acid, formic acid, acetic acid, oxalic acid, succinic acid, tartaric acid, mandelic acid, fumaric acid, maleic acid, lactic acid, citric acid, glutamic acid, saccharic acid, monomethylsebacic acid, 5-oxoproline, hexane-1 -sulphonic acid, nicotinic acid, 2, 3 or 4-aminobenzoic acid, 2,4,6-trimethylbenzoic acid, oc-lipoic acid, acetyl glycine, hippuric acid, phosphoric acid and/or aspartic acid. The free bases of the respective inventive substituted compounds and of corresponding stereoisomers can likewise be converted into the corresponding physiologically acceptable salts using the free acid or a salt of a sugar additive, such as for example saccharin, cyclamate or acesulphame.

Accordingly, the substituted compounds according to the invention such as the free acids of the substituted compounds according to the invention can be converted into the corresponding physiologically acceptable salts by reaction with a suitable base. Examples include the alkali metal salts, alkaline earth metals salts or ammonium salts [NH X R 4 . X ] + , in which x = 0, 1 , 2, 3 or 4 and R represents a branched or unbranched Ci -4 alkyl residue.

The substituted compounds according to the invention and of corresponding stereoisomers can if appropriate, like the corresponding acids, the corresponding bases or salts of these compounds, also be obtained in the form of their solvates, preferably in the form of their hydrates, using conventional methods known to the person skilled in the art.

If the substituted compounds according to the invention are obtained, after preparation thereof, in the form of a mixture of their stereoisomers, preferably in the form of their racemates or other mixtures of their various enantiomers and/or diastereomers, they can be separated and if appropriate isolated using conventional processes known to the person skilled in the art. Examples include chromatographic separating processes, in particular liquid chromatography processes under normal pressure or under elevated pressure, preferably MPLC and HPLC processes, and also fractional crystallisation processes. These processes allow individual enantiomers, for example diastereomeric salts formed by means of chiral stationary phase HPLC or by means of crystallisation with chiral acids, for example (+)- tartaric acid, (-)-tartaric acid or (+)-10-camphorsulphonic acid, to be separated from one another.

The chemicals and reaction components used in the reactions and schemes described below are available commercially or in each case can be prepared by conventional methods known to the person skilled in the art.

General reaction scheme 1 (Scheme 1):

In step j01 an acid halide J-0, in which Hal preferably represents CI or Br, can be esterified using methanol to form the compound J-l by means of methods with which the person skilled in the art is familiar.

In step j02 the methyl pivalate J-l can be converted into an oxoalkylnitrile J-l I by means of methods known to the person skilled in the art, such as for example using acetonitrile CH 3 - CN, if appropriate in the presence of a base.

In step j03 the compound J-ll can be converted into an amino-substituted pyrazolyl derivative J-lll by means of methods known to the person skilled in the art, such as for example using hydrazine hydrate, with cyclization.

In step j04 the amino compound J-lll can first be converted into a diazonium salt by means of methods known to the person skilled in the art, such as for example using nitrite, and the diazonium salt can be converted into a cyano-substituted pyrazolyl derivative J-IV with elimination of nitrogen using a cyanide, if appropriate in the presence of a coupling reagent.

In step j05 the compound J-IV can be substituted in the N position by means of methods known to the person skilled in the art, for example using a halide of part structure (RS2), i.e. Hal-(RS2), if appropriate in the presence of a base and/or a coupling reagent, wherein Hal is preferably CI, Br or I, or using a boronic acid B(OH) 2 (RS2) or a corresponding boronic acid ester, if appropriate in the presence of a coupling reagent and/or a base and the compound J-V can in this way be obtained.

Alternatively, a second synthesis pathway, in which in step k01 an ester K-0 is first reduced to form the aldehyde K-l by means of methods known to the person skilled in the art, for example using suitable hydrogenation reagents such as metal hydrides, is suitable for preparing the compound J-V.

In step k02 the aldehyde K-l can then be reacted with a hydrazine K-V, which can be obtained in step k05, starting from the primary amine K-IV, by means of methods known to the person skilled in the art, to form the hydrazine K-ll by means of methods known to the person skilled in the art with elimination of water.

In step k03 the hydrazine K-ll can be halogenated, preferably chlorinated, by means of methods known to the person skilled in the art with the double bond intact, such as for example using a chlorination reagent such as NCS, and the compound K-lll can in this way be obtained.

In step k04 the hydrazonoyl halide K-lll can be converted into a cyano-substituted compound J-V by means of methods known to the person skilled in the art, such as for example using a halogen-substituted nitrile, with cyclisation.

In step j06 the compound J-V can be hydrogenated by means of methods known to the person skilled in the art, for example using a suitable catalyst such as palladium/activated carbon or using suitable hydrogenation reagents, and the compound (R-ll) can in this way be obtained.

In step j07 the compound (R-ll) can be converted into the compound (R-IV) by means of methods known to the person skilled in the art, such as for example using phenyl chloroformate, if appropriate in the presence of a coupling reagent and/or a base. In addition to the methods disclosed in the present document for preparing unsymmetrical ureas using phenyl chloroformate, there are further processes with which the person skilled in the art is familiar, based on the use of activated carbonic acid derivatives or isocyanates, if appropriate.

In step j08 the amine (R-V) can be converted into the urea compound (R) (wherein A = N). This can be achieved by reaction with (R-IV) by means of methods with which the person skilled in the art is familiar, if appropriate in the presence of a base.

In step j09 the amine (R-ll) can be converted into the amide (R) (wherein A = CH or C(CH 3 )). This can for example be achieved by reaction with an acid halide, preferably a chloride of formula (R-lll) with D = Hal by means of methods with which the person skilled in the art is familiar, if appropriate in the presence of a base or by reaction with an acid of formula (R-lll) with D = OH, if appropriate in the presence of a suitable coupling reagent, for example HATU or CDI, if appropriate with the addition of a base. Further, the amine (R-ll) may be converted into the amide (R) (wherein A = CH or C(CH 3 )) by reaction of a compound (R-llla) by means of methods with which the person skilled in the art is familiar, if appropriate in the presence of a base.

The compounds according to general formula (R), wherein A = N, may be further prepared by a reaction sequence according to general reaction scheme 2. General reaction scheme 2 (scheme 2)

In step v1 the compound (R-V) can be converted into the compound (R-Va) by means of methods known to the person skilled in the art, such as for example using phenyl chloroformate, if appropriate in the presence of a coupling reagent and/or a base. In addition to the methods disclosed in the present document for preparing unsymmetrical ureas using phenyl chloroformate, there are further processes with which the person skilled in the art is familiar, based on the use of activated carbonic acid derivatives or isocyanates, if appropriate.

In step v2 the amine (R-ll) can be converted into the urea compound (R) (wherein A = N). This can be achieved by reaction with (R-Va) by means of methods with which the person skilled in the art is familiar, if appropriate in the presence of a base.

The methods with which the person skilled in the art is familiar for carrying out the reaction steps j01 to j09 and also k01 to k05 as well as v1 and v2 may be inferred from the standard works on organic chemistry such as, for example, J. March, Advanced Organic Chemistry, Wiley & Sons, 6th edition, 2007; F. A. Carey, R. J. Sundberg, Advanced Organic Chemistry, Parts A and B, Springer, 5th edition, 2007; team of authors, Compendium of Organic Synthetic Methods, Wiley & Sons. In addition, further methods and also literature references can be issued by the common databases such as, for example, the Reaxys® database of Elsevier, Amsterdam, NL or the SciFinder® database of the American Chemical Society, Washington, US. EXAMPLES

The following examples further illustrate the invention but are not to be construed as limiting its scope.

The indication ..equivalents" ("eq." or "eq" or "equiv." or "equiv") means molar equivalents, „RT" or "rt" means room temperature (23 ± 7 °C), „M" are indications of concentration in mol/l, „aq." means aqueous, „sat." means saturated, „sol." means solution, "cone." means concentrated.

Further abbreviations: d days

AcOH acetic acid

BH 3 -S(CH 3 ) 2 borane-methyl sulfide complex (BH 3 -DMS)

brine saturated aqueous sodium chloride solution

n-BuLi n-butyllithium

CC column chromatography on silica gel

DBU 1 ,8-diazabicyclo[5.4.0]undec-7-en

DCM dichloromethane

DIPEA diisopropylethylamine

DMA dimethylamine

DMAP 4-dimethylaminopyridine

DMF Ν,Ν-dimethylform amide

EDC N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide

EDCI N-ethyl-N'-(3-dimethylaminopropyl)carbodiimide hydrochloride

ether diethyl ether

EtOAc ethyl acetate

EtOH ethanol

h hour(s)

GC gas chromatography

HBTU 0-(benzotriazol-1 -yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate

HOBt N-hydroxybenzotriazole

H 2 0 water

m/z mass-to-charge ratio

MeOH methanol

MeCN acetonitrile

min or min. minutes MS mass spectrometry

NBS N-bromosuccinamide

TEA triethylamine

NMP N-methyl-2-pyrrolidon

Pd / C palladium on charcoal

Pd 2 (dba) 3 tris(dibenzylideneacetone)dipalladium(0)

Pd(PPh 3 ) 4 tetrakis(triphenylphosphine)palladium(0)

TBTU 0-(benzotriazol-1 -yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate

TLC thin layer chromatography

TFA trifluoroacetic acid

THF tetrahydrofuran

v/v volume to volume

w/w weight in weight

The yields of the compounds prepared were not optimized. All temperatures are uncorrected.

All starting materials which are not explicitly described were either commercially available (the details of suppliers such as for example Acros, Avocado, Aldrich, Apollo, Bachem, Fluka, FluoroChem, Lancaster, Manchester Organics, MatrixScientific, Maybridge, Merck, Rovathin, Sigma, TCI, Oakwood, etc. can be found in the Symyx® Available Chemicals Database of MDL, San Ramon, US or the SciFinder® Database of the ACS, Washington DC, US, respectively, for example) or the synthesis thereof has already been described precisely in the specialist literature (experimental guidelines can be found in the Reaxys® Database of Elsevier, Amsterdam, NL or the SciFinder® Database of the ACS, Washington DC, US, repspectively, for example) or can be prepared using the conventional methods known to the person skilled in the art.

The stationary phase used for the column chromatography was silica gel 60 (0.04 - 0.063 mm) from E. Merck, Darmstadt.

The mixing ratios of solvents or eluents for chromatography are specified in v/v.

All the intermediate products and exemplary compounds were analytically characterized by means of H-NMR spectroscopy. In addition, mass spectrometry tests (MS, m/z for [M+H] + ) were carried out for all the exemplary compounds and selected intermediate products. Synthesis of selected intermediate products:

1. Synthesis of (3-tert-butyl-1 -(3-chlorophenyl)-1 H-pyrazol-5-yl)methanamine (steps i01 -i06)

Step j01 : Pivaloyl chloride (J-0) (1 eq., 60 g) was added dropwise to a solution of methanol (120 mL) within 30 min at 0 'Ό and the mixture was stirred for 1 h at room temperature. After the addition of water (120 mL), the separated organic phase was washed with water (120 mL), dried over sodium sulphate and codistilled with dichloromethane (150 mL). The liquid product J-l was able to be obtained at 99 % purity (57 g).

Step j02: NaH (50 % in paraffin oil) (1 .2 equivalents, 4.6 g) was dissolved in 1 ,4-dioxane (120 mL) and the mixture was stirred for a few minutes. Acetonitrile (1 .2 equivalents, 4.2 g) was added dropwise within 15 min and the mixture was stirred for a further 30 min. The methyl pivalate (J-l) (1 equivalents, 10 g) was added dropwise within 15 min and the reaction mixture was refluxed for 3 h. After complete reaction, the reaction mixture was placed in iced water (200 g), acidified to pH 4.5 and extracted with dichloromethane (12 x 250 mL). The combined organic phases were dried over sodium sulphate, distilled and after recrystallisation from n-hexane (100 mL) 5 g of the product (J-ll) (51 % yield) was able to be obtained as a solid brown substance.

Step j03: At room temperature 4,4-dimethyl-3-oxopentanenitrile (J-ll) (1 equivalents, 5 g) was taken up in ethanol (100 mL), mixed with hydrazine hydrate (2 equivalents, 4.42 g) and refluxed for 3 h. The residue obtained after removal of the ethanol by distillation was taken up in water (100 mL) and extracted with ethyl acetate (300 mL). The combined organic phases were dried over sodium sulphate, the solvent was removed under vacuum and the product (J-ll I ) (5 g, 89 % yield) was obtained as a light red solid after recrystallisation from n- hexane (200 mL).

Step j04: 3-Tert-butyl-1 H-pyrazol-5-amine (J-lll) (1 equivalents, 40 g) was dissolved in diluted HCI (120 mL of HCI in 120 mL of water) and mixed dropwise with NaN0 2 (1 .03 equivalents, 25 g in 100 mL) at 0 - 5 °C over a period of 30 min. After stirring for 30 minutes, the reaction mixture was neutralised with Na 2 C0 3 . A diazonium salt obtained by reaction of KCN (2.4 equivalents, 48 g), water (120 mL) and CuCN (1 .12 equivalents, 31 g) was added dropwise to the reaction mixture within 30 min and the mixture was stirred for a further 30 min at 75 °C. After complete reaction, the reaction mixture was extracted with ethyl acetate (3 x 500 mL), the combined organic phases were dried over sodium sulphate and the solvent was removed under vacuum. The purification (silica gel: 100-200 mesh, eluent: 20 % ethyl acetate/n-hexane) of the residue by column chromatography produced a white solid (J-IV) (6.5 g, 15 %).

Step j05 (method 1):

3-tert.-butyl-1 H-pyrazol-5-carbonitrile (J-IV) (10 mmol) was added to a suspension of NaH (60 %) (12.5 mmol) in dimethylformamide (20 mL) at room temperature while stirring. After stirring for 15 minutes, 1 -iodo-3-chlorobenzene (37.5 mmol) was added dropwise to this reaction mixture at room temperature. After stirring for 30 min at 100 °C, the reaction mixture was mixed with water (150 mL) and extracted with dichloromethane (3 x 75 mL). The combined organic extracts were washed with water (100 mL) and sat. NaCI solution (100 mL) and dried over magnesium sulphate. After removal of the solvent under vacuum, the residue was purified by column chromatography (silica gel: 100-200 mesh, eluent: various mixtures of ethyl acetate and cyclohexane as the mobile solvent) and the product J-V was obtained.

Step j05 [method 2):

A mixture of 3-tert-butyl-1 H-pyrazol-5-carbonitrile (J-IV) (10 mmol), a boronic acid B(OH) 2 (3- chlorophenyl) or a corresponding boronic acid ester (20 mmol) and copper (II) acetate (15 mmol) is placed in dichloromethane (200 mL), mixed with pyridine (20 mmol) while stirring at room temperature and the mixture is stirred for 16 h. After removal of the solvent under vacuum, the residue obtained is purified by column chromatography (silica gel: 100-200 mesh, eluent: various mixtures of ethyl acetate and cyclohexane as the mobile solvent) and the product J-V is in this way obtained.

Step j06: (method 1):

J-V was dissolved together with palladium on carbon (10 %, 500 mg) and concentrated HCI (3 mL) in methanol (30 mL) and exposed to a hydrogen atmosphere for 6 h at room temperature. The reaction mixture was filtered over celite and the filtrate was concentrated under vacuum. The residue was purified by means of flash chromatography (silica gel: 100- 200 mesh, eluent: ethyl acetate) and the product (U-ll) was in this way obtained.

Step j06: (method 2):

J-V was dissolved in tetrahydrofuran (10 mL) and BH 3 » S(CH 3 )2 (2.0 M in tetrahydrofuran, 3 mL, 3 equivalents) was added thereto. The reaction mixture was heated to reflux for 8 h, aq. 2 N HCI (2 N) was added thereto and the reaction mixture was refluxed for a further 30 minutes. The reaction mixture was mixed with aq. NaOH solution (2N) and washed with ethyl acetate. The combined organic phases were washed with sat. aq. NaCI solution and dried over magnesium sulphate. The solvent is removed under vacuum and the residue is purified by column chromatography (silica gel: 100-200 mesh, eluent: various mixtures of dichloromethane and methanol as the mobile solvent) and the product (U-ll) is in this way obtained.

The following further intermediate products were/can be synthesised in a similar manner using the process described hereinbefore under 1.:

(3-tert-butyl-1 -(4-chlorophenyl)-1 H-pyrazol-5-yl)methanamine

(3-tert-butyl-1 -(3-chloro-4-fluorophenyl)-1 H-pyrazol-5-yl)methanamine

(3-tert-butyl-1 -(3-fluorophenyl)-1 H-pyrazol-5-yl)methanamine

(3-tert-butyl-1 -(4-fluorophenyl)-1 H-pyrazol-5-yl)methanamine

(3-tert-butyl-1 -(3,4-difluorophenyl)-1 H-pyrazol-5-yl)methanamine

(3-tert-butyl-1 -(3-methylphenyl)-1 H-pyrazol-5-yl)methanamine

(3-tert-butyl-1 -(3-trifluoromethyl-phenyl)-1 H-pyrazol-5-yl)methanamine

2. Synthesis of 1 -(3-chlorophenyl)-3-(trifluoromethyl)-1 H-pyrazol-5-yl-methanamine (steps k01 -k05 and i06)

Step k01 : LAIH (lithium aluminium hydride) (0.25 equivalents, 0.7g) was dissolved in dry diethyl ether (30 mL) under a protective gas atmosphere and stirred for 2 h at room temperature. The suspension obtained was taken up in diethyl ether (20 mL). Ethyl-2,2,2- trifluoroacetate (K-0) (1 equivalent, 10 g) was taken up in dry diethyl ether (20 mL) and added dropwise to the suspension at -78 'Ό over a period of 1 h. The mixture was then the stirred for a further 2 h at -78 'Ό. ethanol (95 %) (2.5 mL) was then added dropwise, the reaction mixture was heated to room temperature and placed on iced water (30 mL) with concentrated H 2 S0 4 (7.5 mL). The organic phase was separated and concentrated under vacuum and the reaction product K-l was immediately introduced into the next reaction step k02.

Step k05: 3-chloroaniline (K-IV) (1 equivalent, 50 g) was dissolved at -5 to 0 °C in concentrated HCI (300 mL) and stirred for 10 min. A mixture of NaN0 2 (1 .2 equivalents, 32.4 g), water (30 mL), SnCI 2 -2H 2 0 (2.2 equivalents, 70.6 g) and concentrated HCI (100 mL) was added dropwise over a period of 3 h while maintaining the temperature. After stirring for a further 2 h at -5 to 0 'Ό, the reaction mixture was set to pH 9 using NaOH solution and extracted with ethyl acetate (250 mL). The combined organic phases were dried over magnesium sulphate and the solvent was removed under vacuum. The purification by column chromatography (silica gel: 100-200 mesh, eluent: 8 % ethyl acetate/n-hexane) produced 40 g (72 %) of (3-chlorophenyl)hydrazine (K-IV) as a brown oil.

Step k02: The aldehyde (K-l) (2 equivalents, 300 ml_) obtained from k01 and (3- chlorophenyl)hydrazine (K-IV) (1 equivalent, 20 g) were placed in ethanol (200 ml_) and refluxed for 5 h. The solvent was removed under vacuum, the residue was purified by column chromatography (silica gel: 100-200 mesh, eluent: n-hexane) and the product (25 g, 72 %) K-ll was obtained as a brown oil.

Step k03: The hydrazine K-ll (1 equivalent, 25 g) was dissolved in dimethylformamide (125 ml_). N-chlorosuccinimide (1 .3 equivalents, 19.5 g) was added portionwise at room temperature within 15 min and the mixture was stirred for 3 h. The dimethylformamide was removed by distillation and the residue was taken up in ethyl acetate. The ethyl acetate was removed under vacuum, the residue obtained was purified by column chromatography (silica gel: 100-200 mesh, eluent: n-hexane) and the product K-lll (26.5 g, 92 %) was obtained as a pink-coloured oil.

Step k04: At room temperature the hydrazonoyl chloride K-lll (1 equivalent, 10 g) was taken up in toluene (150 ml_) and mixed with 2-chloroacrylonitrile (2 equivalents, 6.1 ml_) and triethylamine (2 equivalents, 10.7 ml_). This reaction mixture was stirred for 20 h at 80 'Ό. The mixture was then diluted with water (200 ml_) and the phases were separated. The organic phase was dried over magnesium sulphate and the solvent was removed under vacuum. The residue was purified by means of column chromatography (silica gel: 100-200 mesh, eluent: 5 % ethyl acetate/n-hexane) and the product (5.5 g, 52 %) was obtained as a white solid J-V.

Step j06 {method 3):

The carbonitrile J-V (1 equivalent, 1 g) was dissolved in methanolic ammonia solution (150 ml_, 1 :1 ) and hydrogenated in an H-cube (10 bar, 80 'Ό, 1 mL/min, 0.25 mol/L). After removal of the solvent under vacuum, (1 -(3-chlorophenyl)-3-(trifluoromethyl)-1 H-pyrazol-5- yl)methanamine (II) was able to be obtained as a white solid (0.92 g, 91 %).

The following further intermediate products were/can be synthesized in a similar manner using the process described hereinbefore under 2.: 1 -(3-fluorophenyl)-3-(trifluoromethyl)-1 H-pyrazol-5-yl)methanamine

1 -(3-chloro-4-fluorophenyl)-3-(trifluoromethyl)-1 H-pyrazol-5-yl)methanamine

1 -(3-methoxyphenyl)-3-(trifluoromethyl)-1 H-pyrazol-5-yl)methanamine

1 -(4-chlorophenyl)-3-(trifluoromethyl)-1 H-pyrazol-5-yl)methanamine

1 -(3,4-difluorophenyl)-3-(trifluoromethyl)-1 H-pyrazol-5-yl)methanamine

1 -(3-trifluoromethyl-phenyl)-3-(trifluoromethyl)-1 H-pyrazol-5-yl)methanamine

1 -(3-methylphenyl)-3-(trifluoromethyl)-1 H-pyrazol-5-yl)methanamine

1 -(3-isopropylphenyl)-3-(trifluoromethyl)-1 H-pyrazol-5-yl)methanamine

3. Preparation of methyl phenyl (3-tert-butyl-1 -(3-chlorophenyl)-1 H-pyrazol-5- vDmethylcarbamate

Step a: To a solution of (3-tert-butyl-1 -(3-chlorophenyl)-1 H-pyrazol-5-yl)methanamine (5 g, 18 mmol) in dimethylformamide (25 mL), potassium carbonate (9.16 g, 66 mmol, 3.5 eq) was added and cooled the contents to O'C. Then phenyl chloroformate (3.28 g (2.65 mL), 20 mmol, 1 .1 equivalents) was added dropwise for 15 minutes and the overall reaction mixture was stirred for another 15 minutes at 0 °C. Progress of the reaction was monitored by TLC (20 % ethyl acetate in n-hexane). On completion of the reaction, reaction contents were filtered, filtrate was diluted with cold water (100 mL) and the product extracted with ethyl acetate (3 χ 25 mL). Combined organic layer was washed with brine solution (100 mL), dried over sodium sulphate and concentrated under reduced pressure. Crude obtained was purified by column chromatography (silica gel: 100-200 mesh, eluent: 10% ethyl acetate in n- hexane) to yield the required product as a white solid (3.2 g, 45 %). 4. Preparation of (1 -(3-chlorophenyl)-3-cvclopropyl-1 H-pyrazol-5-yl)methanamine hydrochloride

Step a: To a solution of sodium ethoxide (freshly prepared by dissolving sodium (1 g, 8.2 mmol, 1 .2 equivalents) in ethanol (30 mL)), diethyl oxalate (0.92 mL, 6.85 mmol, 1 equivalent) was added at room temperature followed by addition of cyclopropyl methyl ketone (0.74 mL, 7.5 mmol, 1 .1 equivalents) dropwise at 0 'Ό. The reaction mixture was slowly warmed to room temperature and stirred for 3 h. Ice cold water (10 mL) was added and ethanol was evaporated under reduced pressure. The residual aqueous layer was diluted with 2 N aq. HCI (15mL) and extracted with diethyl ether (2 χ 25 mL). The organic layer was washed with brine solution and dried over sodium sulphate, filtered and concentrated to give a pale brown liquid (400 mg, 31 %).

Step b: To a solution of step-a product (200 mg, 0.543 mmol, 1 equivalent) in ethanol (8 mL), methoxylamine hydrochloride (30 % solution in water, 0.4 mL, 0.651 mmol, 1 .2 equivalents) was added at room temperature and the reaction mixture stirred for 1 h. ethanol was evaporated under reduced pressure and the residual aqueous layer was extracted with ethyl acetate (15 mL). The organic layer was washed with water (10 mL), brine solution (10 mL), dried over sodium sulphate, filtered and concentrated under reduced pressure to give a pale yellow liquid (180 mg, 78 %).

Step c: A mixture of step-b product (1 .1 g, 5.164 mmol, 1 equivalent) and 3-chlorophenyl hydrazine hydrochloride (1 .84 g, 10.27 mmol, 2 equivalents) was taken in acetic acid (20 mL), 2-methoxy ethanol (10 mL) and the reaction mixture was heated at 105 °C for 3 h. Solvent was evaporated and the residue was extracted with ethyl acetate (60 mL). The organic layer washed with water (10 mL), brine solution (10 mL), dried over sodium sulphate, filtered and concentrated under reduced pressure to give a residue. Purification by column chromatography (silica gel: 100-200 mesh; eluent: ethyl acetate-petroleum ether (4:96)) afforded a pale brown semi solid (1 .15g, 77 %).

Step d: To a solution of step-c product (2.5 g, 8.62 mmol, 1 eq) in tetrahydrofuran (15 mL) - methanol (9 mL) - water (3 mL), lithium hydroxide (1 .08 g, 25.71 mmol, 3 equivalents) was added at 0 °C and the reaction mixture was stirred for 2 h at room temperature. Solvent was evaporated and pH of the residue was adjusted to ~3 sing 2 N aqueous HCI (1 .2 mL). The acidic aqueous layer was extracted with ethyl acetate (2 χ 60 mL); the combined organic layer washed with water (10 mL), brine solution (10 mL), dried over sodium sulphate, filtered and concentrated under reduced pressure to give an off white solid (1 .4 g, 62 %).

Step e: To a solution of step-d product (1 .4 g, 5.34 mmol, 1 equivalent) in 1 ,4-dioxane (30 mL), pyridine (0.25 mL, 3.2 mmol, 0.6 equivalents) and di-tert-butyl dicarbonate (1 .4 mL, 6.37 mmol, 1 .2 equivalents) were added at 0 'C and the resulting mixture was stirred for 30 minutes at the same temperature. Ammonium bicarbonate (0.84 g, 10.63 mmol, 2 equivalents) was added at 0 'C and the reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with water (10 mL) and the aqueous layer was extracted with ethyl acetate (2 χ 30 mL). The organic layer was washed with 2N HCI (20 mL), water (10 mL), brine solution (10 mL), dried over sodium sulphate, filtered and concentrated under reduced pressure to give a residue. Purification by column chromatography (silica gel: 100-200 mesh; eluent: ethyl acetate-petroleum ether (16:84)) gave a white solid (1 g, 72 %).

Step f: To a solution of step-e product (2 g, 7.66 mmol, 1 equivalent) in tetrahydrofuran (25 mL), BH 3 .DMS (1 .44 mL, 15.32 mmol, 2 equivalents) was added at 0 °C and the reaction mixture was heated at 70 ^ for 3 h. The reaction mixture was cooled to 0 'C and methanol (15 mL) was added and reaction mixture heated at reflux for 1 h. The reaction mixture was brought to room temperature and solvent was evaporated under reduced pressure. The residue was dissolved in ether (15 mL), cooled to 0 'C and a solution of HCI in 1 ,4-dioxane (3 mL) was added (pH of the reaction mixture ~4). The precipitated solid was filtered and washed with diethyl ether (5 mL, thrice) to give the hydrochloride salt compound as a white solid (600 mg, 28 %). Synthesis of exemplary compounds:

1. Preparation of amides (A = CH or C(CH?))

General directions for reacting amines of general formula (R-ll) with carboxylic acids of general formula or carboxylic acid derivatives of general formula (R-lll) to form compounds of general formula (R), wherein A = CH or C(CH 3 ) (amides), as in scheme 1 (step j09).

1.1 Method A:

The acid of general formula (R-lll) (1 equivalent), the amine of general formula (R-ll) (1 .2 equivalents) and EDCI (1 .2 equivalents) are stirred in DMF (10 mmol of acid/20 ml_) for 12 hours at RT and water is subsequently added thereto. The reaction mixture is repeatedly extracted with EtOAc, the aqueous phase is saturated with NaCI and subsequently reextracted with EtOAc. The combined organic phases are washed with 1 N HCI and brine, dried over magnesium sulphate and the solvent is removed under vacuum. The residue is purified by means of flash chromatography (Si0 2 , EtOAc/hexane in different ratios such as 1 :2) and the product (R) is in this way obtained.

1.2 Method B:

The acid of general formula (R-lll) (1 equivalent) and the amine of general formulae (R-ll) (1 .1 equivalents) are dissolved in dichloromethane (1 mmol of acid in 6 ml_) and mixed with EDCI (1 .5 equivalents), HOBt (1 .4 equivalents) and triethylamine (3 equivalents) at 0 'C. The reaction mixture is stirred for 20 h at room temperature and the crude product is purified by means of column chromatography (Si0 2 , n-hexane/EtOAc in different ratios such as 2:1 ) and (R) is in this way obtained.

1.3 Method C:

The acid of general formula (R-lll) (1 equivalent) is first mixed with a chlorinating agent, preferably with thionyl chloride and the mixture obtained in this way is boiled under reflux and the acid (R-lll) is in this way converted into the corresponding acid chloride. The amine of general formulae (R-ll) (1 .1 equivalents) is dissolved in dichloromethane (1 mmol of acid in 6 ml_) and mixed with triethylamine (3 equivalents) at 0 'C. The reaction mixture is stirred for 20 h at room temperature and the crude product is purified by means of column chromatography (Si0 2 , n-hexane/EtOAc in different ratios such as 2:1 ) and (R) is in this way obtained.

1.4 Method D:

The phenyl ester (R-llla) (1 equivalent) and the corresponding amine (R-ll) (1 .1 equivalents) are dissolved in THF (10 mmol of the reaction mixture in 120 mL) and stirred for 16 h at room temperature after addition of DBU (1 .5 equivalents). After removal of the solvent under vacuum, the residue obtained is purified by means of flash chromatography (Si0 2 , EtOAc/hexane in different ratios such as 1 :1 ) and (R) is in this way obtained.

The exemplary compounds B1 -B10, B13, B15-B23, B26, B29-B30, B37, B42-B43, B47, BBS- Bee, B76, B82, B88, B93-B98 and B102-B104 and B106-B107 were obtained using one of the methods described hereinbefore.

2. Preparation of ureas (A = N)

General directions for reacting amines of general formula (R-ll) or (R-V) with phenyl chloroformate to form compounds of formula (R-IV) or (R-Va) (scheme 1 , step j07 and scheme 2, step v1 ) and subsequent reaction of compounds of formula (R-IV) with amines of general formula (R-V) (scheme 1 , step j08) or of compounds of formula (R-Va) with amines of general formula (R-ll) (scheme 2, step v2) to form compounds of general formula (R), wherein A = N:

Step j07/step v1 : The amine of general formula (R-ll) or (R-V) (1 equivalent) is placed in dichloromethane (10 mmol of amine in 70 mL) and phenyl chloroformate (1 .1 equivalents) is added thereto at room temperature and the mixture is stirred for 30 min. After removal of the solvent under vacuum, the residue is purified by means of flash chromatography (Si0 2 , diethyl ether/hexane in different ratios such as 1 :2) and (R-IV) or (R-Va) is in this way obtained.

Step j08/step v2: The carbamic acid phenyl ester (R-IV) or (R-Va) obtained (1 equivalent) and the corresponding amine (R-V) or (R-ll) (1 .1 equivalents) are dissolved in THF (10 mmol of the reaction mixture in 120 mL) and stirred for 16 h at room temperature after addition of DBU (1 .5 equivalents). After removal of the solvent under vacuum, the residue obtained is purified by means of flash chromatography (Si0 2 , EtOAc/hexane in different ratios such as 1 :1 ) and (R) is in this way obtained. The exemplary compounds B11-B12, B15, B24-B25, B27-B28, B31-B36, B38-B41, B44-B46, B48-B62, B69-B75, B77-B81, B83-B87, B89-B92 and B99-B101 as well as B105 were obtained using one of the methods described hereinbefore..

Detailed synthesis of selected exemplary compounds

Synthesis of example B5: N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[4-(methanesulfonamido-methyl)-phenyl]-propionamid e

Step 1 : To a stirred solution of (4-bromophenyl)methanamine (500 mg, 2.687 mmol) in pyridine were added methanesulfonyl chloride (0.4 ml_, 5.106 mmol) at O'C. The reaction mixture was stirred for 1 h, then diluted with dichloromethane. The mixture was washed with water. The organic layer was dried (MgS0 4 ) and filtered. The solvent removed in vacuo. The crude was purified by CC. N-(4-bromobenzyl)methanesulfonamide (675 mg) was obtained (95 % yield).

Step 2: To a stirred solution of N-(4-bromobenzyl)methanesulfonamide (675 mg, 2.555 mmol) in DMF were added ethyl 2-chloropropionate (0.42 ml_), manganese (280 mg) and (2,2'-bipyridine)nickel(ll)-dibromide (NiBr 2 bipy) (67 mg, 0.17885mmol). TFA (1 -2 drops) was added. The reaction mixture was stirred for 36 h at 60 °C. After cooling down to room temperature, the mixture was hydrolyzed by 1 N HCI and extracted with diethyl ether. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by CC. Ethyl 2-(4-(methylsulfonamidomethyl)phenyl)propanoate (325 mg) was obtained.

Step 3: To a stirred solution of ethyl 2-(4-(methylsulfonamidomethyl)phenyl)propanoate (325 mg, 1 .139 mmol) in a co-solvent of THF and water (1 :1 ) was added sodium hydroxide (1 14 mg, 2.8475 mmol). The reaction mixture was refluxed for 16 h, then cooled to room temperature, acidified to pH 3-4 with AcOH. The residue dissolved in EtOAc and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent removed in vacuo. The crude was purified by CC. 2-(4-(methylsulfonamidomethyl)phenyl)propanoic acid (74 mg) was obtained in 25 % yield.

Step 4: To a stirred solution of 2-(4-(methylsulfonamidomethyl)phenyl)propanoic acid (37 mg, 0.144 mmol) and (1 -(3-chlorophenyl)-3-(trifluoromethyl)-1 H-pyrazol-5-yl)methanamine (44 mg, 0.158 mmol) in acetonitrile were added EDC (41 mg, 0.216 mmol), HOBt (29 mg, 0.216 mmol) and triethylamine (0.05 mL, 0.36 mmol). The reaction mixture was stirred for 15 h at room temperature. The residue dissolved in EtOAc and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by CC. Example compound B5 (62 mg) was obtained in 84 % yield. H NMR (300 MHz, CDCI 3 ): δ 7.35 (m, 8H), 6.35 (s, 1 H), 5.56 (t, 1 H), 4.5 (m, 3H), 4.32 (d, 2H), 3.53 (q, 1 H), 2.94 (s, 3H), 1 .50 (d, 3H).

Synthesis of example B17: N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[4-[[(ethylsulfonyl)amino]-methyl]-3-fluoro-phenyl ]-propionamide

Step 1 : 4-Bromo-2-fluorobenzylamine (924 mg, 4.53 mmol) was dissolved in pyridine and ethane sulfonyl chloride (0.82 mL, 8.60 mmol) was added to the solution at O 'C. The mixture was stirred for 1 h at O 'C. Then, the mixture was quenched with 1 N HCI and extracted with ethyl acetate (EtOAc). Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by CC (eluent EtOAc/n-hexane) to yield N-(4-bromo-2- fluorobenzyl)ethanesulfonamide in pure form (1 .06 g, 79 %).

Step 2: To a solution of N-(4-bromo-2-fluorobenzyl)ethanesulfonamide (305 mg, 1 .03 mmol) in DMF, manganese (1 13 mg, 2.06 mmol), NiBr 2 bipy (27 mg, 0.07 mmol), ethyl-2-chloro propionate (0.17 ml, 1 .34 mmol) was added, followed by addition of TFA (0.002 ml, 0.028 mmol). The mixture was stirred for 1 day at 65 ° C. The reaction mixture was quenched by cone. HCI (7-drops) and then extracted with diethyl ether, dried (MgS0 4 ) and the solvent was evaporated in vacuo. The residue was purified by CC (EtOAc/n-hexane). Ethyl 2-(4- (ethylsulfonamidomethyl)-3-fluorophenyl)propanoate in pure form (65 mg , 20 %) was obtained.

Step 3: To a solution of ethyl 2-(4-(ethylsulfonamidomethyl)-3-fluorophenyl)propanoate (305 mg, 1 .03 mmol) in DMF, manganese (1 13 mg, 2.06 mmol), NiBr 2 bipy (27 mg, 0.07 mmol), ethyl-2-chloro propionate (0.17 mL, 1 .34 mmol) was added, followed by addition of TFA (0.002 mL, 0.028 mmol). The mixture was stirred for 1 day at 65 ° C. The reaction mixture was quenched by cone. HCI (7-drops) and then extracted with diethyl ether, dried (MgS0 4 ) and the solvent was evaporated in vacuo. The residue was purified by CC (EtOAc/n-hexane). 2-(4-(ethyl-sulfonamidomethyl)-3-fluorophenyl)propanoic acid in pure form (65 mg, 20 %) was obtained.

Step 4: 2-(4-(ethyl-sulfonamidomethyl)-3-fluorophenyl)propanoic acid (60 mg, 0.207 mmol) and (1 -(3-chlorophenyl)-3-(trifluoromethyl)-1 H-pyrazol-5-yl)methanamine (63 mg, 0.228 mmol) were dissolved and mixed in 1 ,4-dioxane followed by addition of HOBt (42 mg, 0.310 mmol) and EDC (60 mg, 0.313 mmol) and TEA (0.07 mL, 0.518 mmol). The reaction mixture was stirred for overnight and then quenched by water and extracted with EtOAc. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by CC (EtOAc/n-hexane) to give the example B17 in pure form (104 mg, 92 %).

H-NMR (CD 3 OD): δ 7.50 (m, 5 H, Ar), 7.05 (m, 2 H, Ar), 6.57 (s, 1 H, Ar), 4.43 (m, 2 H, Ar- CH 2 ), 4.27 (s, 2 H, Ar-CH 2 ), 3.58 (q, 1 H , J = 7.14 Hz, amide 1 H), 2.97 (q, 2 H, J = 7.32 Hz, ethanesulfonylamide 2 H), 1 .36 (d , 3 H, J = 7.14 Hz, amide 3 H), 1 .27 (t, 3 H, J = 7.32 Hz, ethanesulfonylamide 3 H).

Synthesis of example B22: 2-[3-Chloro-4-(methanesulfonamido-methyl)-phenyl]-N-[[2-(3- chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-p ropionamide

Step 1 : To a stirred solution of 1 (3 g, 16.078 mmol) in methanol (35 mL) was added sulfuric acid (0.3 mL). The reaction mixture was refluxed for 15 h and cooled to room temperature. The solvent was evaporated. The residue was dissolved in EtOAc and extracted with a sat. solution of NaHC0 3 . The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by CC. 2 (3.557 g) was obtained in 99 % yield.

Step 2: To a stirred solution of 2 (3.557 g, 17.73 mmol) and TEA (2.5 mL, 17.73 mmol) in dichloromethane, triflic anhydride (3 mL, 17.73 mmol) is added dropwise at O 'C. The reaction mixture was stirred for 2 h. The residue was extracted with CH 2 CI 2 and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by CC. 3 (5.15 g) was obtained in 87 % yield. Step 3: To a stirred solution of 3 (4.419 g, 13.283 mmol) in DMF were added zinc(ll) cyanide (1 .6 g, 13.681 mmol) and Pd(PPh 3 ) 4 (1 .5 g, 1 .3283 mmol). The reaction mixture was stirred for 34 hours at 80 °C, then cooled to room temperature and diluted with EtOAc. The mixture was filtered using a celite pad. The filtrate was diluted with EtOAc and extracted with a sat. solution of NaHC0 3 . The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 4 (1 .044 g) was obtained in 37 % yield.

Step 4: To a stirred solution of 4 (931 mg, 4.441 mmol) in DMF were added sodium hydride (60 wt.-% in mineral oil, 178 mg, 4.441 mmol) and iodomethane (0.3 ml, 4.441 mmol) at 0 ° C. The reaction mixture was stirred for 1 hour at Ο'Ό, then diluted with water. The residue dissolved in EtOAc and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 5 (642 mg) was obtained in 65 % yield.

Step 5: To a stirred solution of 5 (642 mg, 2.870 mmol) in a co-solvent of THF and water (1 :1 ) was added sodium hydroxide (287 mg, 7.175 mmol). The reaction mixture was stirred for 15 hours at room temperature, then acidified to pH 3-4 with AcOH. The residue was dissolved in EtOAc and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 6 (665 mg) was obtained in 99 % yield.

Step 6: To a stirred solution of 6 (224 mg, 1 .069 mmol) and 7 (324 mg, 1 .175 mmol) in acetonitrile were added EDC (307 mg, 1 .064 mmol), HOBt (217 mg, 1 .064 mmol) and triethylamine (0.4 ml_, 2.673 mmol). The reaction mixture was stirred for 15 hours at room temperature. The mixture was diluted with EtOAc and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 8 (366 mg) was obtained in 78 % yield.

Step 7: To a stirred solution of 8 (366 mg, 1 .460 mmol) in methanol, cooled to 0°C, were added Boc 2 0 (342 mg, 1 .566 mmol) and NiCI 2 -6H 2 0 (19 mg, 0.0783 mmol). NaBH 4 (207 mg, 5.481 mmol) was then added in small portions. The reaction was exothermic and

effervescent. The resulting reaction mixture was allowed to warm to room temperature and left to stir for 1 hour. Diethylenetriamine (DETA) (0.09 ml_, 0.783 mmol) was added to the mixture. The mixture was stirred for 1 hour. The solvent was evaporated. The residue dissolved in EtOAc and extracted with a sat. solution of NaHC0 3 . The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 9 (227 mg) was obtained in 50 % yield.

Step 8: To a stirred solution of 9 (227 mg, 0.397 mmol) in dichloromethane (4 mL), cooled to 0°C, were added trifluoroacetic acid (2 mL). The resulting reaction mixture was stirred for 1 hour at 0°C and 1 hour at room temperature, then basified to pH 8-9 with aq. NaHC0 3 . The mixture was filtered using a celite pad. The filtrate was dissolved in dichloromethane and extracted with a sat. solution of NaHC0 3 . The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 10 (1 16 mg) was obtained in 62 % yield.

Step 9: To a stirred solution of 10 (1 16 mg, 0.246 mmol) in pyridine, cooled to Ο 'Ό, was added methanesulfonyl chloride (1 16 mg). The resulting reaction mixture was stirred for 15 hours at room temperature. The mixture dissolved in dichloromethane and washed with 1 N HCI. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 11 (108 mg) was obtained as 80 % yield. H NMR (300 MHz, CDCI 3 ): δ 7.43 (m, 4H), 7.29 (m, 3H), 7.15 (dd, 1 H, J=7.86 Hz), 6.43 (s, 1 H), 5.63 (t, 1 H), 4.76 (t, 1 H), 4.48 (d, 2H), 4.40 (d, 2H), 3.48 (q, 1 H, J=7.14 Hz), 2.90 (s, 3H), 1 .47 (d, 3H, J=7.14 Hz).

Synthesis of example B28: 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[4- (methanesulfonamido-methyl)-phenyl]-urea

Step 1 : To a stirred solution of 1 (299 mg, 1 .952 mmol) in dichloromethane was added triethylamine (0.3 ml, 2.147 mmol). Methanesulfonyl chloride (0.18 ml, 2.343 mmol) is added dropwise at Ο 'Ό. The reaction mixture was heated to 80 °C and stirred for 4 hours, then cooled to room temperature, and diluted with dichloromethane. The mixture was washed with water. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 2 (333 mg) was obtained in 74 % yield.

Step 2: To a stirred solution of 2 (333 mg, 1 .440 mmol) in DMF was added potassium phthallimide (293 mg, 1 .584 mmol). The reaction mixture was stirred for 16 hours. The mixture was dissolved in EtOAc, washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent removed in vacuo. The crude was purified by column chromatography. 3 (535 mg) was obtained as a crude.

Step 3: To a stirred solution of 3 (218 mg, 0.772 mmol) in THF were added hydrazine monohydrate (246 mg, 3.089 mmol) and p-toluenesulfonic acid monohydrate (15 mg, 0.0772 mmol). The reaction mixture was stirred for 4 hours at δΟ 'Ό, then cooled to room

temperature, and diluted with EtOAc. The mixture was washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 4 (46 mg) was obtained in 39 % yield.

Step 4: To a stirred solution of 4 (46 mg, 0.302 mmol) in pyridine, cooled to 0 ° C, was added methanesulfonyl chloride (46 mg). The resulting reaction mixture was stirred for 1 hour at room temperature. The mixture dissolved in dichloromethane and washed with 1 N HCI. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 5 (43 mg) was obtained in 62 % yield.

Step 5: To a stirred solution of 5 (43 mg, 0.188 mmol) in EtOAc was added 10 % palladium on carbon (5 mg). The mixture was charged with a H 2 (gas) balloon. The resulting mixture was stirred for 3 hours, then filtered over celite. The solvent was removed in vacuo. The crude was purified by column chromatography. 6 (41 mg) was obtained in 99 % yield.

Step 6: To a stirred solution of 6 (41 mg, 0.204 mmol) in tetrahydrofuran and acetonitrile as co-solvent were added phenylchloroformate (34 mg, 0.2142 mmol) and pyridine (0.02 ml_, 0.2448 mmol). The reaction mixture was stirred for 3 hours at room temperature. The mixture was diluted with EtOAc and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 7 (54 mg) was obtained in 83 % yield.

Step 7: To a stirred solution of 7 (28 mg, 0.087 mmol) and 8 (23 mg, 0.087 mmol) in acetonitrile was added DMAP (1 1 mg, 0.087 mmol). The reaction mixture was stirred for 15 hours at 50 °C. The mixture was diluted with EtOAc and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 9 (example compound B28) (32 mg) was obtained in 75 % yield.

H NMR (400 MHz, CDCI 3 ): δ 7.43 (s, 1 H), 7.30 (m, 2H), 7.12 (q, 4H), 6.89 (s, 1 H), 6.24 (s, 1 H), 5.28 (d, 1 H), 4.92 (t, 1 H), 4.39 (d, 2H), 4.15 (d, 2H), 4.09 (q, 1 H), 2.85 (s, 3H), 1 .28 (s, 9H). Synthesis of example B29: N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[3-fluoro-4-[(methyl-methylsulfonyl-amino)-methyl] -phenyl]-propionam

Step 1 : Commercially available (4-bromo-2-fluorophenyl)methanamine is stirred in pyridine and methanesulfonyl chloride (1 .9 eq.) is added drop-wise at 0°C. The reaction mixture is stirred at room-temperature for 1 h. The reaction is quenched with 1 N HCI and extracted with EtOAc. The organic layer is dried over by MgS0 4 and the solvent evaporated. The crude is purified by column chromatography and 1 obtained.

Step 2: Compound 1 is dissolved in anhydrous DMF and charged with N 2 . Commercially available rthyl-2-chloro propionate (1 .3 eq.) is added dropwise and manganese (2 eq.), NiBr 2 bipy (0.1 equiv.) are added followed by TFA (0.026 eq.). The reaction mixture is refluxed overnight. The reaction mixture is warmed to ambient temperature. The reaction is quenched with 1 N HCI and the organic layer is extracted with diethyl ether. The extracted organic layer is dried over MgS0 4 , and concentrated into compound 2 that is used in the next step without further purification.

Step 3: Crude compound 2 is stirred in acetone at 0°C, and K 2 C0 3 (1 .5 eq.) is added. Methyl iodide (3 eq.) is added drop-wise and the reaction mixture is refluxed. After 15 h, the reaction mixture is quenched with water and extracted with EtOAc. The organic layer is dried over MgS0 4 and concentrated. The crude is purified by column chromatography and obtained as desired product 3.

Step 4: To a solution of compound 3 in THF and water (1 :1 ) was added NaOH (2.5 equiv.) and the resulting mixture was stirred at room temperature. After 15 hours, the reaction mixture was acidified with AcOH acid to a pH of 2-3. The mixture is extracted with DCM and water. The organic layer was washed with water, dried (MgS0 4 ) and concentrated in vacuo. The product was purified by column chromatography and the desired product 4 was obtained.

Step 5: To a solution of the carboxylic acid (4) in 1 ,4-dioxane was added EDC (1 .5 eq.), HOBt (1 .5 eq.), and (1 -(3-chlorophenyl)-3-(trifluoromethyl)-1 H-pyrazol-5-yl)methanamine (1 eq.) and dropwise TEA (2.5 eq.). The reaction mixture was stirred overnight at room temperature. Water was added to the reaction mixture and the mixture was extracted with ethyl acetate. The extracted organic layer was dried over MgS0 4 . Evaporation of the solvent followed by column chromatographic purification (EtOAc/n-hexane) afforded example compound B29.

H NMR (300 MHz, CDCI 3 ): δ 7.41 -7.32 (m, 5H, Ar-H), 6.99-6.95 (m, 3H, Ar-H), 6.06 (s, 1 H, Ar-H), 5.643 (bs, 1 H, Ar-NH), 4.46 (d, 2H, J=6.00Hz, pyrazole-a-H), 3.88 (s, 2H, Ar-a-H), 3.49 (q, 1 H, J=7.50Hz, Ar-a-H), 2.48 (s, 3H, methansulfonyl-CH 3 ), 1 .47 (d, 3H, J=6.00Hz, Ar- a-CH 3 )

Synthesis of example B31 : 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[3- fluoro-4-(methanesulfonamido-methyl)-phenyl]-urea

Step 1 : To a stirred solution of 1 (1.993 g, 12.847 mmol) in carbon tetrachloride were added benzoyl peroxide (497 mg, 1 .2847 mmol) and N-bromosuccinimide (2.972 g, 16.701 mmol). The reaction mixture was refluxed for 18 h, then cooled to room temperature. The mixture was diluted with EtOAc, then washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 2 (780 mg) was obtained in 26 % yield.

Step 2: To a stirred solution of 2 (780 mg, 3.333 mmol) in DMF was added potassium phthallimide (1 .235 g, 6.666 mmol). The reaction mixture was stirred for 18 h. The mixture was dissolved in EtOAc, washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 3 (1 .034 g) was obtained as crude.

Step 3: To a stirred solution of 3 (1.034 g, 3.444 mmol) in THF were added hydrazine monohydrate (1 .104 g, 13.776 mmol) and p-toluenesulfonic acid monohydrate (66 mg, 0.3444 mmol). The reaction mixture was refluxed for 6 hours, then cooled to room

temperature, and diluted with EtOAc. The mixture was washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 4 (329 mg) was obtained in 56 % yield.

Step 4: To a stirred solution of 4 (131 mg, 0.770 mmol) in pyridine, cooled to 0°C, was added methanesulfonyl chloride (131 mg). The resulting reaction mixture was stirred for 1 hour at room temperature. The mixture was diluted with dichloromethane and washed with 1 N HCI. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 5 (173 mg) was obtained in 91 % yield.

Step 5: To a stirred solution of 5 (187 mg, 0.753 mmol) in tetrahydrofuran and ethanol as co- solvent were added 10 % palladium on carbon (20 mg). The mixture was charged with H 2 (gas) balloon. The resulting mixture was stirred for 15 hours, then filtered using celite. The solvent was removed in vacuo. The crude was purified by column chromatography. 6 (135 mg) was obtained in 82 % yield.

Step 6: To a stirred solution of 6 (135 mg, 0.618 mmol) in tetrahydrofuran and acetonitrile as co-solvent were added phenylchloroformate (0.08 ml_, 0.6489 mmol) and pyridine (0.06 ml_, 0.7416 mmol). The reaction mixture was stirred for 1 hour at room temperature. The mixture was diluted with EtOAc and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 7 (140 mg) was obtained in 67 % yield.

Step 7: To a stirred solution of 7 (46 mg, 0.136 mmol) and 8 (36 mg, 0.136 mmol) in acetonitrile was added DMAP (17 mg, 0.136 mmol). The reaction mixture was stirred for 15 hours at 50 ° C. The mixture was diluted with EtOAc and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 9 (72 mg) was obtained in 99 % yield.

H NMR (300 MHz, DMSO-d 6 ): δ 8.84 (s, 1 H), 7.60 (s, 1 H), 7.45 (m, 4H), 7.26 (t, 1 H), 7.01 (dd, 1 H), 6.76 (t, 1 H), 6.32 (s, 1 H), 4.40 (d, 2H), 4.09 (d, 2H), 2.85 (s, 3H), 1 .27 (s, 9H). Synthesis of example B35: 1 -[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-3-[4-[(sulfamoylamino)-methyl]-phenyl]-urea

10

Step 1 : NBS (1 .51 g, 8.509 mmol) was added to a solution of 4-nitro-toluene 1 (1 .2 g, 7.735 mmol) in carbon tetrachloride. 70% Benzoyl peroxide (120 mg) was added to the mixture at room temperature. The mixture was refluxed. After 24 h, the mixture was extracted with ethyl acetate (EtOAc). Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 2 in pure form in 61 % yield.

Step 2: To a solution of compound 2 (1 .1 g , 4.69 mmol) in DMF, potassium phthalimide(1 .9 g, 10.314 mmol) was added. The mixture was stirred overnight and then extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 3 in pure form in 99 % yield.

Step 3: To a solution of compound 3 (1 .6 g, 5.33 mmol) in THF, hydrazine monohydrate (4 eq) was added. The mixture was refluxed for 6 hours and cooled to RT. The mixture was treated with potassium bicarbonate to a pH of 12-13. The mixture was extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 4 in pure form in 65 % yield (592 mg).

Step 4: Chlorosulfonyl isocyanate (0.063 ml_) and t-BuOH (0.07 ml_) were mixed in DCM. After 10 minutes, a solution of compound 4 (100 mg, 0.657 mmol) in DCM was added at 50 'C. After stirring for 30 min. the mixture was cooled to room temperature and then TEA (0.1 1 ml_) was added and the mixture was stirred for 3 hours and then extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 5 in pure form in 51 % yield (1 12 mg).

Step 5: 10% Palladium on carbon (7 mg) was added to a solution of compound 5 (65 mg) in ethanol and THF and the mixture was charged with H 2 (g). After stirring the reaction mixture for 6 h, the mixture was filtered using Celite and the solvent evaporated in vacuo to give the compound 6 in pure form in 58 % yield (98 mg).

Step 6: Compound 6 (86 mg, 0.285 mmol) was dissolved in THF/acetonitrile. Pyridine (0.03 ml_, 0.342 mmol) was added and then it addition of phenylchloroformate (0.04 ml, 0.300 mmol) at O 'C followed. The mixture was stirred at 0°C for 30 min and heated up to room temperature and then it was stirred for 30 min. After that, it was extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 7 in pure form in 49 % yield (59 mg).

Step 7: Compound 7 (58 mg, 0.138 mmol) was dissolved in MeCN. Compound 8 (38 mg, 0.138 mmol) and DMAP (16 mg) were added to the solution. The reaction mixture was stirred overnight at 50 °C. The mixture was extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 9 in pure form in 50 % yield (60 mg).

Step 8: To a solution of compound 9 (80 mg, 0.133 mmol) in DCM (6 ml_), TFA (2 ml_) is added at 0 °C. The mixture was stirred for 30 min and stirred for 2 hr more at room temperature. The mixture was neutralized with sodium bicarbonate to a pH of 7-8 and then extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 10 in pure form in 75 % yield (50 mg).

H-NMR(CD 3 OD): 7.64 (m, 1 H, Ar), 7.55 (m, 3 H, Ar), 7.28 (m, 4 H, Ar), 6.75 (s, 1 H, Ar), 4.47 (s, 2 H, CH 2 NH), 4.09 (s, 2 H , CH 2 NH).

Synthesis of example B40: 1 -[4-(Aminomethyl)-3-fluoro-phenyl]-3-[[5-tert-butyl-2-(3- chlorophenyl)-2H-pyrazol-3-yl]-methyl]-urea

10

Steps 1-3 are performed as described for the synthesis of example B31

Step 4: Compound 4 (100 mg, 0.588 mmol) was dissolved in DCM. At 0°C, Boc 2 0 (154 mg, 0.705 mmol) was added to the solution. After stirring for 30 min, the mixture was heated to room temperature and then TEA (0.13 ml) was added and the mixture was stirred overnight. And then it was extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 5 in pure form in 86 % yield (136 mg). Step 5: 10% Palladium on carbon (20 mg) was added to a solution of compound 5 (136 mg) in ethanol and THF and the mixture was charged with H 2 (g). After stirring the reaction mixture for 6 h, the mixture was filtered using Celite and the solvent was evaporated in vacuo. Compound 6 was obtained in 85% (103 mg).

Step 6: Compound 6 (103 mg, 0.429 mmol) was dissolved in THF/MeCN. Pyridine (0.04 mL, 0.515 mmol) was added and then addition of phenylchloroformate (0.06 mL, 0.450 mmol) at O 'C was followed. The mixture was stirred at 0°C for 30 min and heated up to room temperature and then it was stirred for another 30 min. After that, it was extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 7 in pure form in 75 % yield (1 16 mg).

Step 7: Compound 7 (70 mg, 0.194 mmol) was dissolved in MeCN. Compound 8 (52 mg, 0.137 mmol) and DMAP (24 mg) were added to the solution. The reaction mixture was stirred overnight at 50 °C. The mixture was extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 9 in pure form in 97 % yield (100 mg).

Step 8: To a solution of compound 9 (100 mg, 0.189 mmol) in DCM (6 mL), TFA (2 mL) is added at O 'C and the mixture was stirred for 30 min and stirred for 2 h more at room temperature. The mixture was neutralized with sodium bicarbonate to a pH of 7-8 and then extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 10 in pure form in 74 % yield (60 mg).

H-NMR (300 MHz, DMSO-d6): δ 9.00 (s, 1 H, urea), 7.60 (s, 1 H, Ar), 7.47 (m, 4 H, Ar, urea), 7.33 (m, 1 H, Ar), 7.07 (m, 1 H, Ar), 6.94 (m, 1 H, Ar), 6.31 (s, 1 H, Ar), 4.39 (m, 2 H, Ar-CH 2 ), 3.88 (s, 2 H, Ar-CH 2 ), 1 .26 (s, 9 H.t-butyl 9 H). Synthesis of example B46: 1 -[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl]- 3-[3- fluoro-4-[(sulfamoylamino)-methyl]-phenyl]-urea

10

Steps 1-3 are performed as described for the synthesis of example B31

Step 4: Chlorosulfonyl isocyanate (0.1 mL) and t-BuOH (0.12 mL) were mixed in DCM. After 10 minutes, a solution of compound 4 (200 mg, 1 .176 mmol) in DCM was added at 50 'C. After stirring for 30 min. the mixture was cooled to room temperature and then TEA (0.1 1 mL) was added and the mixture was stirred for 3 hours and then extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 5 in pure form in 23 % yield (139 mg).

Step 5: 10% Palladium on carbon (42 mg) was added to a solution of compound 5 (135 mg) in ethanol and THF and the mixture was charged with H 2 (g). After stirring the reaction mixture for 6 h, the mixture was filtered using Celite and the solvent evaporated in vacuo to give the compound 6 in pure form in 99 % yield (127 mg).

Step 6: Compound 6 (127 mg, 0.398 mmol) was dissolved in THF/acetonitrile. Pyridine (0.04 ml, 0.478 mmol) was added and then addition of phenylchloroformate (0.05 mL, 0.418 mmol) at O 'C followed. The mixture was stirred at O'C for 30 min and heated up to room

temperature and then it was stirred for 30 min. After that, it was extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 7 in pure form in 91 % yield (160 mg).

Step 7: Compound 7 (50 mg, 0.1 14 mmol) was dissolved in MeCN. Compound 8 (30 mg, 0.1 14 mmol) and DMAP (14 mg) were added to the solution. The reaction mixture was stirred overnight at 50 °C. The mixture was extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 9 in pure form in 65 % yield (45 mg).

Step 8: To a solution of compound 9 (45 mg, 0.074 mmol) in DCM (6 mL), TFA (2 mL) is added at 0 °C. The mixture was stirred for 30 min and stirred for 2 h more at room

temperature. The mixture was neutralized with sodium bicarbonate to a pH of 7-8 and then extracted with EtOAc and washed by brine. Drying (MgS0 4 ) and evaporation of the ethyl acetate followed and the residue was purified by column chromatography (EtOAc/n-hexane) to give the compound 10 in pure form in 74 % yield (28 mg).

H-NMR (300 MHz, CD 3 OD): δ 7.48 (m, 4 H, Ar), 7.33 (m, 2 H, Ar), 6.96 (m, 1 H, Ar), 6.36 (s, 1 H, Ar), 4.41 (s, 2 H), 4.17 (s, 2 H), 1 .32 (s, 9 H, t-butyl 9 H).

Synthesis of example B63: N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[3- fluoro-4-(methylamino-methyl)-phenyl]-propionamide

8

Step 1 : To a stirred solution of 2-(3-Fluoro-4-nitro-phenyl)-propionic acid in methanol was added sulfuric acid (0.3 mL). The reaction mixture was refluxed for 15 h and cooled to room temperature. The solvent was evaporated. The residue was dissolved in EtOAc and extracted with a sat. solution of NaHC0 3 . The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by CC. 1 was obtained.

Step 2: 10% Palladium on carbon was added to a solution of ethyl ester (1 ) in ethanol and THF and the mixture was charged with H 2 (g). After stirring the reaction mixture for 6 h, the mixture was filtered using Celite and purified by column chromatography to obtain 2. Step 3: A solution of p-TsOH.H 2 0 (3 equiv.) in CH 3 CN was added to a solution of 2 (1 eq.) in CH 3 CN. The resulting suspension was cooled to 10-15 °C and to this was added, gradually, a solution of NaN0 2 (2 eq.) and Kl (2.5 eq.) in H 2 0. The reaction mixture was stirred for 10 min then allowed to come to 20 °C and stirred until the starting material was consumed. After 4 hours, water, NaHC0 3 (until pH=9-10) was added and extracted with ethyl acetate. The organic layer was washed with water, dried (MgS0 4 ) and concentrated in vacuo. The residue was purified by CC to give 3.

Step 4: Compound 3, Pd 2 (dba) 3 , dppf, Zn powder, Zn(CN) 2 were placed in round flask charged with N 2 and DMA (0.02 equiv.) was added dropwise by syringe. The reaction mixture was stirred at 120^ for 15 hours and cooled to room temperature followed by extraction with EtOAc and washing with a 2N NH 4 OH solution. The organic layer was washed with water, dried (Na 2 S0 4 ) and concentrated in vacuo. The residue was purified on CC to give the desired product 4.

Step 5: To a solution of compound 4 in THF and water (1 :1 ) is added NaOH (2.5 equiv.) and the mixture stirred at room temperature. After 15 hours, the reaction mixture is acidified by AcOH until pH = 2-3. The mixture is extracted with DCM and water. The organic layer is washed with water, dried (MgS0 4 ) and concentrated in vacuo. The product is purified by CC (eluent DCM:MeOH 10:1 ) and the desired product 5 obtained.

Step 6: EDC (1 .5 equiv), HOBt (1 .5 equiv), and (3-tert-butyl-1 -(3-chlorophenyl)-1 H-pyrazol-5- yl)methanamine (1 eq.) are added to a solution of (5) and then added drop-wise is TEA (2.5 ml_). The reaction mixture is stirred overnight at room temperature. The reaction is quenched with water and extracted with ethyl acetate. The extracted organic layer is dried over MgS0 4 . After evaporation of the solvent, the residue is purified by column chromatographic purification (eluent EtOAc:n-hexane) and 6 is obtained.

Step 7: Nickel(ll) chloride hexahydrate (1 eq.) and compound 6 are stirred in anhydrous ethanol during 15 minute for activation. Sodium borohydride (7 eq.) is added the mixture stirred for 2 h. Celite was added to the reaction and it was filtered by using a celite packed filter, and washing with ethanol was performe. The residue is purified after concentration to obtain 7.

Step 8: Sodium methoxide (1 M in methanol) was added to a solution of compound 7 in methanol followed by the addition of paraformaldehyde (5 eq.). The reaction mixture was refluxed for 1 .5 h then cooled to O'C with an ice bath. Sodium borohydride (6 equiv.) was added with caution. The mixture was refluxed again for 1 h and cooled down. The mixture was extracted with DCM and washed with water. After evaporating the solvent, the mixture was purified by column chromatography (DCM:MeOH = 4:1 ) to obtain example compound B63.

H NMR (300 MHz, CDCI 3 ): δ 7.41 -7.32 (m, 5H, Ar-H), 6.99-6.95 (m, 3H, Ar-H), 6.06 (s, 1 H, Ar-H), 5.643 (bs, 1 H, Ar-NH), 4.46 (d, 2H, J=6.00Hz, pyrazole-a-H), 3.88 (s, 2H, Ar-a-H), 3.49 (q, 1 H, J=7.50Hz, Ar-a-H), 2.48 (s, 3H, methansulfonyl-CH 3 ), 1 .47 (d, 3H, J=6.00Hz, Ar- a-CH 3 ), 1 .33 (s, 9H, t-butyl).

Synthesis of example B64: N-[[5-tert-Butyl-2-(3-chlorophenyl)-2H-pyrazol-3-yl]-methyl] -2-[4- (dimethylaminomethyl)-3-fluoro-phenyl]-propionamide

Steps 1-7 are performed as described for the synthesis of example B63

Step 8: Sodium cyanoborohydrid is added to a solution of compound 7 and

paraformaldehyde in acetic acid. The reaction mixture is stirred at room temperature for 15 hours and poured on ice. Sodium bicarbonate was used to adjust the pH to 9. The mixture was extracted with EtOAc and dried over sodium sulfate. After evaporating solvent, the mixture is purified by column chromatography to obtain the desired product example compound B64.

H NMR (400 MHz, CDCI 3 ): δ 7.38 (s, 1 H, Ar-H), 7.35-7.27 (m, 3H, Ar-H), 7.21 (d, 1 H, Ar-H), 6.95 (m, 2H, Ar-H), 6.01 (s, 1 H, Ar-H), 5.48 (bs, 1 H, Ar-NH), 4.46 (d, 2H, J=6.00Hz, pyrazole- a-H), 3.68 (s, 2H, Ar-a-H), 3.49 (q, 1 H, J=7.50Hz, Ar-a-H), 2.30 (s, 6H, methansulfonyl-CH 3 and N-CH 3 ), 1 .47 (d, 3H, J=6.00Hz, Ar-a-CH 3 ), 1 .33 (s, 9H, t-butyl).

Synthesis of example B86: 1 -[4-(Methanesulfonamido-methyl)-3-methoxy-phenyl]-3-[[2-(m- tolyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]-methyl]-urea

7

Step 1 : To a stirred solution of 1 (300 mg, 1 .683 mmol) in THF were added borane methyl sulfide complex (2M in THF) (1 .4 mL, 2.83 mmol) at room temperature. The reaction mixture was stirred for 16 h at 66 °C, then cooled to room temperature. The residue was diluted with EtOAc and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 2 (270 mg) was obtained as 68 % yield.

Step 2: To a stirred solution of 2 (190 mg, 1 .04 mmol) in pyridine was added

methanesulfonyl chloride at 0 ° C. The reaction mixture was stirred for 1 h at room

temperature. The mixture was quenched by 1 N HCI. The residue was diluted with DCM and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 3 (205 mg) was obtained as 76 % yield.

Step 3: To a stirred solution of 3 (205 mg, 0.79 mmol) in tetrahydrofuran and ethanol as co- solvent was added 10 % palladium on carbon (21 mg). The mixture was charged with H 2 (gas) balloon. The resulting mixture was stirred for 16 h, then filtered using celite. The filtrate removed in vacuo. The filtrate removed in vacuo. The crude was purified by column chromatography. 4 (190 mg) was obtained as 99 % yield.

Step 4: To a stirred solution of 4 (190 mg, 0.83 mmol) in THF (6 ml_) and CH 3 CN (8 ml_) as co-solvent were added phenyl chloroformate (0.1 1 ml_, 0.866 mmol) and pyridine (0.08 ml_, 0.99 mmol). The reaction mixture was stirred for 3 h at room temperature. The residue was dissolved in EtOAc and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 5 (238 mg) was obtained as 82 % yield.

Step 5: To a stirred solution of 5 (79 mg, 0.225 mmol) in MeCN and 6 (57 mg, 0.225 mmol) were added DMAP (28 mg, 0.225 mmol). The reaction mixture was stirred for 16 hours at 50 'Ό. The residue dissolved in EtOAc and washed with water and brine. The organic layer was dried (MgS0 4 ) and filtered. The solvent was removed in vacuo. The crude was purified by column chromatography. 7 (104 mg) was obtained as 90 % yield.

H NMR (300 MHz, DMSO) 57.42 (m, 4H, Ar), 7.17 (m, 2H, Ar), 6.83(d, J=9.87 Hz, 1 H, Ar), 6.77 (s, 1 H, Ar), 4.38 (d, 2H, J=5.67 Hz, CH 2 ), 4.03 (d, 2H, J=6.06 Hz, CH 2 ), 3.74 (s, 3H, methoxy), 2.81 (s, 3H, mesyl), 2.40 (s, 3H, Ar-CH 3 ). Synthesis of example B95: N-[[2-(3-Chlorophenyl)-5-cyclopropyl-2H-pyrazol-3-yl]-methyl ]- 2-[3-fluoro-4-[(sulfamoylamino)-methyl]-phenyl]-propionamide

Steps 1-5 are performed as described for the synthesis of example B63

Step 6: EDC (1 .5 equiv), HOBt (1 .5 equiv), and (1 -(3-chlorophenyl)-3-cyclopropyl-1 H- pyrazol-5-yl)methanamine (1 eq.) are added to a solution of (5) and then added drop-wise is TEA (2.5 mL). The reaction mixture is stirred overnight at room temperature. The reaction is quenched with water and extracted with ethyl acetate. The extracted organic layer is dried over MgS0 4 . After evaporation of the solvent, the residue is purified by column

chromatographic purification (eluent EtOAc/n-hexane) and 6 is obtained. Step 7: Nickel(ll) chloride hexahydrate (1 eq.) and compound 6 are stirred in anhydrous ethanol during 15 minute for activation. Sodium borohydride (7 eq.) is added the mixture stirred for 2 hours. Celite was added to the reaction and it was filtered by using a celite packed filter, and washing with ethanol was performed. The residue is purified after concentration to obtain 7.

Step 8: Compound 7 and compound 8 are dissolved in DCM. TEA (0.1 eq.) is added dropwise. The reaction mixture is stirred for 15 h at room temperature and quenched with water. The organic layer is extracted with DCM and concentrated. After purification by CC, compound 9 is obtained.

Step 9: TFA (12 ml_) is added to a solution of compound 9 in a solution of DCM and the reaction mixture is stirred for 4 h at room temperature. Water is added to the mixture and the separated mixture is extracted with DCM. The residue is purified after concentration by CC and compound 10 (example B95) is obtained.

H NMR (400 MHz, CDCI 3 ) δ 7.41 -7.33 (m, 5H, Ar-H), 6.94 (m, 2H, Ar-H), 5.39 (s, 1 H, Ar-H), 5.39 (bs, 1 H, a-NH), 4.43 (bs, 1 H, Ar-a-NH), 4.46 (2, 2H, Ar-a-CH 2 ), 4.41 (m, 2H, J=6.00Hz, a-CH 2 ), 4.32 (d, 2H, a-CH 2 ), 3.47 (m, 1 H), 2.38 (s, 3H, Ar-CH 3 ), 1 .44 (d, 3H, J=6.00Hz, a- CH 3 ), 0.93 (m, 2H, cyclopropyl-CH 2 ), 0.68 (m, 2H, cyclopropyl-CH 2 ).

Step 10: 1 eq. of CSI (Chlorosulfonyl isocyanate) is added dropwise to a cold solution of tert- butyl alcohol (1 eq.) in anhydrous DCM. Then DMAP (2 eq.) is added. The mixture is stirred for 3 h at room temperature. The organic layer is extracted with DCM and washed with water. After column chromatography, a colorless powder (compound 8) is obtained.

Synthesis of example B97: N-[[2-(3-Chlorophenyl)-5-(trifluoromethyl)-2H-pyrazol-3-yl]- methyl]-2-[3-fluoro-4-[(sulfamoylamino)-methyl]-phenyl]-prop ionamide

10

Steps 1-5 are performed as described for the synthesis of example B95

Step 6: EDC (1 .5 equiv), HOBt (1 .5 equiv), and (1 -(3-chlorophenyl)-3-(trifluoromethyl)-1 H- pyrazol-5-yl)methanamine (1 eq.) are added to a solution of (5) and then added drop-wise is TEA (2.5 mL). The reaction mixture is stirred overnight at room temperature. The reaction is quenched with water and extracted with ethyl acetate. The extracted organic layer is dried over MgS0 4 . After evaporation of the solvent, the residue is purified by column

chromatographic purification (eluent EtOAc/n-hexane) and 6 is obtained. Steps 7-10 are performed as described for the synthesis of example B95

NMR characterization of example B97 (compound 10):

H NMR (400 MHz, CDCI 3 ): δ 7.41 -7.33 (m, 5H, Ar-H), 6.94 (m, 2H, Ar-H), 6.40 (s, 1 H, Ar-H), 5.59 (bs, 1 H, a-NH), 4.64 (bs, 1 H, Ar-a-NH), 4.46 (s, 2H, Ar-a-CH 2 ), 4.32 (d, 2H, J=8.00 Hz, a-CH 2 ), 4.09 (q, 1 H, a-CH), 1 .44 (d, 3H, J=6.00Hz, a-CH 3 )

Mass spectrometric data are cited hereinafter by way of example for the following exemplary compounds (Table 1 ):

Table 1

Exemplary [M+H] Exemplary [M+H] Exemplary [M+H] compound compound compound

B1 533.2 B47 442.5 B93 522.7

B2 550.8 B48 504.0 B94 542.0

B3 538.9 B49 521 .9 B95 506.5

B4 489.9 B50 492.6 B96 540.6

B5 515.4 B51 526.8 B97 534.5

B6 503.6 B52 520.6 B98 533.9

B7 517.0 B53 492.0 B99 478.1

B8 567.0 B54 542.1 B100 510.2

B9 505.1 B55 554.0 B101 484.1

B10 523.0 B56 475.0 B102 516.8

B11 520.5 B57 505.1 B103 504.6

B12 532.4 B58 515.0 B104 529.3

B13 538.0 B59 493.1 B105 517.6

B14 438.2 B60 504.9 B106 529.3

B15 529.2 B61 493.3 B107 517.6

B16 535.6 B62 522.9

B17 547.5 B63 457.7

B18 533.4 B64 471 .5

B19 535.0 B65 483.6

B20 521 .6 B66 497.4

B21 505.2 B67 485.6

B22 549.9 B68 513.2

B23 519.0 B69 51 1 .0

B24 470.0 B70 539.4

B25 458.1 B71 517.0

B26 507.2 B72 510.1

B27 538.1 B73 528.1

B28 490.6 B74 492.9

B29 547.5 B75 475.3

B30 535.6 B76 545.5

B31 508.6 B77 486.9

B32 526.0 B78 521 .6

B33 484.2 B79 51 1 .0

B34 472.5 B80 529.1

B35 503.6 B81 502.4

B36 491 .5 B82 541 .1

B37 503.0 B83 510.0

B38 470.0 B84 516.7

B39 442.2 B85 504.6

B40 430.6 B86 51 1 .9

B41 521 .4 B87 500.1

B42 455.3 B88 501 .2

B43 555.0 B89 481 .9

B44 485.9 B90 470.2

B45 474.0 B91 495.9

B46 509.7 B92 484.1 Pharmacological methods

I. Functional testing carried out on the vanilloid receptor 1 (VRI/TRPV1 receptor)

The agonistic or antagonistic effect of the substances to be tested on the rat-species vanilloid receptor 1 (VR1/TRPV1 ) can be determined using the following assay. In this assay, the influx of Ca 2+ through the receptor channel is quantified with the aid of a Ca 2+ -sensitive dye (type Fluo-4, Molecular Probes Europe BV, Leiden, the Netherlands) in a fluorescent imaging plate reader (FLIPR, Molecular Devices, Sunnyvale, USA).

Method:

Complete medium: 50 ml_ HAMS F12 nutrient mixture (Gibco Invitrogen GmbH, Karlsruhe, Germany) with 10 % by volume of FCS (foetal calf serum, Gibco Invitrogen GmbH, Karlsruhe, Germany, heat-inactivated); 2mM L-glutamine (Sigma, Munich, Germany); 1 % by weight of AA solution (antibiotic/antimyotic solution, PAA, Pasching, Austria) and 25 ng/mL NGF medium (2.5 S, Gibco Invitrogen GmbH, Karlsruhe, Germany)

Cell culture plate: Poly-D-lysine-coated, black 96-well plates having a clear base (96-well black/clear plate, BD Biosciences, Heidelberg, Germany) are additionally coated with laminin (Gibco Invitrogen GmbH, Karlsruhe, Germany), the laminin being diluted with PBS (Ca-Mg- free PBS, Gibco Invitrogen GmbH, Karlsruhe, Germany) to a concentration of 100 μg/mL. Aliquots having a laminin concentration of 100 μg mL are removed and stored at -20 'C. The aliquots are diluted with PBS in a ratio of 1 : 10 to 10 μg mL of laminin and respectively 50 μ\- of the solution are pipetted into a recess in the cell culture plate. The cell culture plates are incubated for at least two hours at 37 °C, the excess solution is removed by suction and the recesses are each washed twice with PBS. The coated cell culture plates are stored with excess PBS which is not removed until just before the feeding of the cells.

Preparation of the cells:

The vertebral column is removed from decapitated rats and placed immediately into cold HBSS buffer (Hank's buffered saline solution, Gibco Invitrogen GmbH, Karlsruhe, Germany), i.e. buffer located in an ice bath, mixed with 1 % by volume (per cent by volume) of an AA solution (antibiotic/antimyotic solution, PAA, Pasching, Austria). The vertebral column is cut longitudinally and removed together with fasciae from the vertebral canal. Subsequently, the dorsal root ganglia (DRG) are removed and again stored in cold HBSS buffer mixed with 1 % by volume of an AA solution. The DRG, from which all blood remnants and spinal nerves have been removed, are transferred in each case to 500 μΙ_ of cold type 2 collagenase (PAA, Pasching, Austria) and incubated for 35 minutes at 37 <Ό. After the addition of 2.5 % by volume of trypsin (PAA, Pasching, Austria), incubation is continued for 10 minutes at 37 <Ό. After complete incubation, the enzyme solution is carefully pipetted off and 500 μΙ_ of complete medium are added to each of the remaining DRG. The DRG are respectively suspended several times, drawn through cannulae No. 1 , No. 12 and No. 1 6 using a syringe and transferred to a 50 ml_ Falcon tube which is filled up to 15 ml_ with complete medium. The contents of each Falcon tube are respectively filtered through a 70 μηι Falcon filter element and centrifuged for 1 0 minutes at 1 ,200 rpm and room temperature. The resulting pellet is respectively taken up in 250 μΙ_ of complete medium and the cell count is determined.

The number of cells in the suspension is set to 3 x 1 0 5 per ml_ and 1 50 μΙ_ of this suspension are in each case introduced into a recess in the cell culture plates coated as described hereinbefore. In the incubator the plates are left for two to three days at 37 °C, 5 % by volume of C0 2 and 95 % relative humidity. Subsequently, the cells are loaded with 2 μΜ of Fluo-4 and 0.01 % by volume of Pluronic F1 27 (Molecular Probes Europe BV, Leiden, the Netherlands) in HBSS buffer (Hank's buffered saline solution, Gibco Invitrogen GmbH, Karlsruhe, Germany) for 30 min at 37 °C, washed 3 times with HBSS buffer and after further incubation for 1 5 minutes at room temperature used for Ca 2+ measurement in a FLIPR assay. The Ca 2+ -dependent fluorescence is in this case measured before and after the addition of substances ( ex = 488 nm, em = 540 nm). Quantification is carried out by measuring the highest fluorescence intensity (FC, fluorescence counts) over time.

FLIPR assay:

The FLIPR protocol consists of 2 substance additions. First the compounds to be tested (10 μΜ) are pipetted onto the cells and the Ca 2+ influx is compared with the control (capsaicin 1 0 μΜ). This provides the result in % activation based on the Ca 2+ signal after the addition of 1 0 μΜ of capsaicin (CP). After 5 minutes' incubation, 100 nM of capsaicin are applied and the Ca 2+ influx is also determined.

Desensitising agonists and antagonists lead to suppression of the Ca 2+ influx. The % inhibition is calculated compared to the maximum achievable inhibition with 1 0 μΜ of capsazepine. Triple analyses (n=3) are carried out and repeated in at least 3 independent experiments (N=4).

Starting from the percentage displacement caused by different concentrations of the compounds to be tested of general formula I, IC 5 o inhibitory concentrations which cause a 50- % displacement of capsaicin were calculated. K values for the test substances were obtained by conversion by means of the Cheng-Prusoff equation (Cheng, Prusoff; Biochem. Pharmacol. 22, 3099-3108, 1973).

II. Functional testing carried out on the vanilloid receptor 1 (VRI/TRPV1 receptor)

The agonistic or antagonistic effect of the substances to be tested on the vanilloid receptor 1 (VR1 ) can also be determined using the following assay. In this assay, the influx of Ca 2+ through the channel is quantified with the aid of a Ca 2+ -sensitive dye (type Fluo-4, Molecular Probes Europe BV, Leiden, the Netherlands) in a fluorescent imaging plate reader (FLIPR, Molecular Devices, Sunnyvale, USA).

Method:

Chinese hamster ovary cells (CHO K1 cells, European Collection of Cell Cultures (ECACC) United Kingdom) are stably transfected with the VR1 gene. For functional testing, these cells are plated out on poly-D-lysine-coated black 96-well plates having a clear base (BD Biosciences, Heidelberg, Germany) at a density of 25,000 cells/well. The cells are incubated overnight at 37 °C and 5 % C0 2 in a culture medium (Ham's F12 nutrient mixture, 1 0 % by volume of FCS (foetal calf serum), 1 8 μg mL of L-proline). The next day the cells are incubated with Fluo-4 (Fluo-4 2 μΜ, 0.01 % by volume of Pluronic F1 27, Molecular Probes in HBSS (Hank's buffered saline solution), Gibco Invitrogen GmbH, Karlsruhe, Germany) for 30 minutes at 37 <C. Subsequently, the plates are washed three times with HBSS buffer and after further incubation for 15 minutes at RT used for Ca 2+ measurement in a FLIPR assay. The Ca 2+ -dependent fluorescence is measured before and after the addition of the substances to be tested ( ex wavelength = 488 nm, e m = 540 nm). Quantification is carried out by measuring the highest fluorescence intensity (FC, fluorescence counts) over time. FLIPR assay:

The FLIPR protocol consists of 2 substance additions. First the compounds to be tested (10 μΜ) are pipetted onto the cells and the Ca 2+ influx is compared with the control (capsaicin 10 μΜ) (% activation based on the Ca 2+ signal after the addition of 10 μΜ of capsaicin). After 5 minutes' incubation, 100 nM of capsaicin are applied and the Ca 2+ influx is also determined.

Desensitising agonists and antagonists led to suppression of the Ca 2+ influx. The % inhibition is calculated compared to the maximum achievable inhibition with 10 μΜ of capsazepine.

Starting from the percentage displacement caused by different concentrations of the compounds to be tested of general formula I, IC 50 inhibitory concentrations which cause a 50- per cent displacement of capsaicin were calculated. K, values for the test substances were obtained by conversion by means of the Cheng-Prusoff equation (Cheng, Prusoff; Biochem. Pharmacol. 22, 3099-3108, 1973).

Pharmacological data

The affinity of the compounds according to the invention for the vanilloid receptor 1 (VR1/TRPV1 receptor) was determined as described hereinbefore (pharmacological method I or II).

The compounds according to the invention display outstanding affinity to the VR1 /TRPV1 receptor (Table 2).

In Table 2 the abbreviations below have the following meanings:

Cap = capsaicin

AG = agonist

The value after the „@"symbol indicates the concentration at which the inhibition (as a percentage) was respectively determined.

Table 2